1
|
Eng JR, Bucher E, Hu Z, Walker CR, Risom T, Angelo M, Gonzalez-Ericsson P, Sanders ME, Chakravarthy AB, Pietenpol JA, Gibbs SL, Sears RC, Chin K. Highly multiplexed imaging reveals prognostic immune and stromal spatial biomarkers in breast cancer. JCI Insight 2025; 10:e176749. [PMID: 39808504 PMCID: PMC11948582 DOI: 10.1172/jci.insight.176749] [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: 10/17/2023] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Spatial profiling of tissues promises to elucidate tumor-microenvironment interactions and generate prognostic and predictive biomarkers. We analyzed single-cell spatial data from 3 multiplex imaging technologies: cyclic immunofluorescence (CycIF) data we generated from 102 patients with breast cancer with clinical follow-up as well as publicly available mass cytometry and multiplex ion-beam imaging datasets. Similar single-cell phenotyping results across imaging platforms enabled combined analysis of epithelial phenotypes to delineate prognostic subtypes among patients who are estrogen-receptor+ (ER+). We utilized discovery and validation cohorts to identify biomarkers with prognostic value. Increased lymphocyte infiltration was independently associated with longer survival in triple-negative (TN) and high-proliferation ER+ breast tumors. An assessment of 10 spatial analysis methods revealed robust spatial biomarkers. In ER+ disease, quiescent stromal cells close to tumor were abundant in tumors with good prognoses, while tumor cell neighborhoods containing mixed fibroblast phenotypes were enriched in poor-prognosis tumors. In TN disease, macrophage/tumor and B/T lymphocyte neighbors were enriched, and lymphocytes were dispersed in good-prognosis tumors, while tumor cell neighborhoods containing vimentin+ fibroblasts were enriched in poor-prognosis tumors. In conclusion, we generated comparable single-cell spatial proteomic data from several clinical cohorts to enable prognostic spatial biomarker identification and validation.
Collapse
Affiliation(s)
- Jennifer R. Eng
- Department of Molecular and Medical Genetics and
- Department of Biomedical Engineering, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | - Elmar Bucher
- Department of Biomedical Engineering, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | - Zhi Hu
- Department of Biomedical Engineering, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | - Cameron R. Walker
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | - Tyler Risom
- Department of Research Pathology, Genentech, South San Francisco, California, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA
| | | | - Melinda E. Sanders
- Vanderbilt-Ingram Cancer Center and
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center (VUMC), Nashville, Tennessee, USA
| | | | - Jennifer A. Pietenpol
- Vanderbilt-Ingram Cancer Center and
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA
| | - Summer L. Gibbs
- Department of Biomedical Engineering, Oregon Health and Science University (OHSU), Portland, Oregon, USA
| | | | - Koei Chin
- Department of Biomedical Engineering, Oregon Health and Science University (OHSU), Portland, Oregon, USA
- Cancer Early Detection Advanced Research Center, OHSU, Portland, Oregon, USA
| |
Collapse
|
2
|
Miyazawa K, Itoh Y, Fu H, Miyazono K. Receptor-activated transcription factors and beyond: multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem 2024; 300:107256. [PMID: 38569937 PMCID: PMC11063908 DOI: 10.1016/j.jbc.2024.107256] [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: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine that is widely distributed throughout the body. Its receptor proteins, TGF-β type I and type II receptors, are also ubiquitously expressed. Therefore, the regulation of various signaling outputs in a context-dependent manner is a critical issue in this field. Smad proteins were originally identified as signal-activated transcription factors similar to signal transducer and activator of transcription proteins. Smads are activated by serine phosphorylation mediated by intrinsic receptor dual specificity kinases of the TGF-β family, indicating that Smads are receptor-restricted effector molecules downstream of ligands of the TGF-β family. Smad proteins have other functions in addition to transcriptional regulation, including post-transcriptional regulation of micro-RNA processing, pre-mRNA splicing, and m6A methylation. Recent technical advances have identified a novel landscape of Smad-dependent signal transduction, including regulation of mitochondrial function without involving regulation of gene expression. Therefore, Smad proteins are receptor-activated transcription factors and also act as intracellular signaling modulators with multiple modes of function. In this review, we discuss the role of Smad proteins as receptor-activated transcription factors and beyond. We also describe the functional differences between Smad2 and Smad3, two receptor-activated Smad proteins downstream of TGF-β, activin, myostatin, growth and differentiation factor (GDF) 11, and Nodal.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hao Fu
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory for Cancer Invasion and Metastasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
Xu M, Zhang T, Xia R, Wei Y, Wei X. Targeting the tumor stroma for cancer therapy. Mol Cancer 2022; 21:208. [PMID: 36324128 PMCID: PMC9628074 DOI: 10.1186/s12943-022-01670-1] [Citation(s) in RCA: 135] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Tumors are comprised of both cancer cells and surrounding stromal components. As an essential part of the tumor microenvironment, the tumor stroma is highly dynamic, heterogeneous and commonly tumor-type specific, and it mainly includes noncellular compositions such as the extracellular matrix and the unique cancer-associated vascular system as well as a wide variety of cellular components including activated cancer-associated fibroblasts, mesenchymal stromal cells, pericytes. All these elements operate with each other in a coordinated fashion and collectively promote cancer initiation, progression, metastasis and therapeutic resistance. Over the past few decades, numerous studies have been conducted to study the interaction and crosstalk between stromal components and neoplastic cells. Meanwhile, we have also witnessed an exponential increase in the investigation and recognition of the critical roles of tumor stroma in solid tumors. A series of clinical trials targeting the tumor stroma have been launched continually. In this review, we introduce and discuss current advances in the understanding of various stromal elements and their roles in cancers. We also elaborate on potential novel approaches for tumor-stroma-based therapeutic targeting, with the aim to promote the leap from bench to bedside.
Collapse
Affiliation(s)
- Maosen Xu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Tao Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Ruolan Xia
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, West China Hospital, National Clinical Research Center for Geriatrics, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041, Chengdu, Sichuan, PR China.
| |
Collapse
|
4
|
Abstract
Despite the decline in death rate from breast cancer and recent advances in targeted therapies and combinations for the treatment of metastatic disease, metastatic breast cancer remains the second leading cause of cancer-associated death in U.S. women. The invasion-metastasis cascade involves a number of steps and multitudes of proteins and signaling molecules. The pathways include invasion, intravasation, circulation, extravasation, infiltration into a distant site to form a metastatic niche, and micrometastasis formation in a new environment. Each of these processes is regulated by changes in gene expression. Noncoding RNAs including microRNAs (miRNAs) are involved in breast cancer tumorigenesis, progression, and metastasis by post-transcriptional regulation of target gene expression. miRNAs can stimulate oncogenesis (oncomiRs), inhibit tumor growth (tumor suppressors or miRsupps), and regulate gene targets in metastasis (metastamiRs). The goal of this review is to summarize some of the key miRNAs that regulate genes and pathways involved in metastatic breast cancer with an emphasis on estrogen receptor α (ERα+) breast cancer. We reviewed the identity, regulation, human breast tumor expression, and reported prognostic significance of miRNAs that have been documented to directly target key genes in pathways, including epithelial-to-mesenchymal transition (EMT) contributing to the metastatic cascade. We critically evaluated the evidence for metastamiRs and their targets and miRNA regulation of metastasis suppressor genes in breast cancer progression and metastasis. It is clear that our understanding of miRNA regulation of targets in metastasis is incomplete.
Collapse
Affiliation(s)
- Belinda J Petri
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Carolyn M Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
5
|
Bathula NV, Bommadevara H, Hayes JM. Nanobodies: The Future of Antibody-Based Immune Therapeutics. Cancer Biother Radiopharm 2020; 36:109-122. [PMID: 32936001 DOI: 10.1089/cbr.2020.3941] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Targeted therapy is a fast evolving treatment strategy to reduce unwanted damage to healthy tissues, while increasing efficacy and specificity. Driven by state-of-the-art technology, this therapeutic approach is especially true of cancer. Antibodies with their remarkable specificity have revolutionized therapeutic strategies for autoimmune conditions and cancer, particularly blood-borne cancers, but have severe limitations in treating solid tumors. This is mainly due to their large molecular size, low stability, tumor-tissue penetration difficulties, and pharmacokinetic properties. The tumor microenvironment, rich in immune-suppressing molecules is also a major barrier in targeting solid tumors by antibody-based drugs. Nanobodies have recently emerged as an alternative therapeutic agent to overcome some of the drawbacks of traditional antibody treatment. Nanobodies are the VHH domains found on the heavy-chain only antibodies of camelids and are the smallest naturally available antibody fragments with excellent antigen-binding specificity and affinity, equivalent to conventional antibodies but with molecular weights as low as 15 kDa. The compact size, high stability, enhanced hydrophilicity, particularly in framework regions, excellent epitope interactions with protruding CDR3 regions, and improved tissue penetration make nanobodies the next-generation therapeutics (Nano-BioDrugs). In this review, the authors discuss the interesting properties of nanobodies and their advantages over their conventional counterparts and provide insight into how nanobodies are being utilized as agonists and antagonists, bispecific constructs, and drug and enzyme-conjugates to combat the tumor microenvironment and treat disease.
Collapse
Affiliation(s)
- Nuthan V Bathula
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Hemashree Bommadevara
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Jerrard M Hayes
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
Gaponova AV, Rodin S, Mazina AA, Volchkov PV. Epithelial-Mesenchymal Transition: Role in Cancer Progression and the Perspectives of Antitumor Treatment. Acta Naturae 2020; 12:4-23. [PMID: 33173593 PMCID: PMC7604894 DOI: 10.32607/actanaturae.11010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022] Open
Abstract
About 90% of all malignant tumors are of epithelial nature. The epithelial tissue is characterized by a close interconnection between cells through cell-cell interactions, as well as a tight connection with the basement membrane, which is responsible for cell polarity. These interactions strictly determine the location of epithelial cells within the body and are seemingly in conflict with the metastatic potential that many cancers possess (the main criteria for highly malignant tumors). Tumor dissemination into vital organs is one of the primary causes of death in patients with cancer. Tumor dissemination is based on the so-called epithelial-mesenchymal transition (EMT), a process when epithelial cells are transformed into mesenchymal cells possessing high mobility and migration potential. More and more studies elucidating the role of the EMT in metastasis and other aspects of tumor progression are published each year, thus forming a promising field of cancer research. In this review, we examine the most recent data on the intracellular and extracellular molecular mechanisms that activate EMT and the role they play in various aspects of tumor progression, such as metastasis, apoptotic resistance, and immune evasion, aspects that have usually been attributed exclusively to cancer stem cells (CSCs). In conclusion, we provide a detailed review of the approved and promising drugs for cancer therapy that target the components of the EMT signaling pathways.
Collapse
Affiliation(s)
- A. V. Gaponova
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - S. Rodin
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, 17177 Sweden
| | - A. A. Mazina
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| | - P. V. Volchkov
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, 141701 Russia
| |
Collapse
|
7
|
Gibbs ZA, Reza LC, Cheng CC, Westcott JM, McGlynn K, Whitehurst AW. The testis protein ZNF165 is a SMAD3 cofactor that coordinates oncogenic TGFβ signaling in triple-negative breast cancer. eLife 2020; 9:57679. [PMID: 32515734 PMCID: PMC7302877 DOI: 10.7554/elife.57679] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/09/2020] [Indexed: 12/19/2022] Open
Abstract
Cancer/testis (CT) antigens are proteins whose expression is normally restricted to germ cells yet aberrantly activated in tumors, where their functions remain relatively cryptic. Here we report that ZNF165, a CT antigen frequently expressed in triple-negative breast cancer (TNBC), associates with SMAD3 to modulate transcription of transforming growth factor β (TGFβ)-dependent genes and thereby promote growth and survival of human TNBC cells. In addition, we identify the KRAB zinc finger protein, ZNF446, and its associated tripartite motif protein, TRIM27, as obligate components of the ZNF165-SMAD3 complex that also support tumor cell viability. Importantly, we find that TRIM27 alone is necessary for ZNF165 transcriptional activity and is required for TNBC tumor growth in vivo using an orthotopic xenograft model in immunocompromised mice. Our findings indicate that aberrant expression of a testis-specific transcription factor is sufficient to co-opt somatic transcriptional machinery to drive a pro-tumorigenic gene expression program in TNBC.
Collapse
Affiliation(s)
- Zane A Gibbs
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
| | - Luis C Reza
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
| | - Chun-Chun Cheng
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
| | - Jill M Westcott
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kathleen McGlynn
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
| | - Angelique W Whitehurst
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, United States.,Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
8
|
Yang Y, Yang HH, Tang B, Wu AML, Flanders KC, Moshkovich N, Weinberg DS, Welsh MA, Weng J, Ochoa HJ, Hu TY, Herrmann MA, Chen J, Edmondson EF, Simpson RM, Liu F, Liu H, Lee MP, Wakefield LM. The Outcome of TGFβ Antagonism in Metastatic Breast Cancer Models In Vivo Reflects a Complex Balance between Tumor-Suppressive and Proprogression Activities of TGFβ. Clin Cancer Res 2019; 26:643-656. [PMID: 31582516 DOI: 10.1158/1078-0432.ccr-19-2370] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/28/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022]
Abstract
PURPOSE TGFβs are overexpressed in many advanced cancers and promote cancer progression through mechanisms that include suppression of immunosurveillance. Multiple strategies to antagonize the TGFβ pathway are in early-phase oncology trials. However, TGFβs also have tumor-suppressive activities early in tumorigenesis, and the extent to which these might be retained in advanced disease has not been fully explored. EXPERIMENTAL DESIGN A panel of 12 immunocompetent mouse allograft models of metastatic breast cancer was tested for the effect of neutralizing anti-TGFβ antibodies on lung metastatic burden. Extensive correlative biology analyses were performed to assess potential predictive biomarkers and probe underlying mechanisms. RESULTS Heterogeneous responses to anti-TGFβ treatment were observed, with 5 of 12 models (42%) showing suppression of metastasis, 4 of 12 (33%) showing no response, and 3 of 12 (25%) showing an undesirable stimulation (up to 9-fold) of metastasis. Inhibition of metastasis was immune-dependent, whereas stimulation of metastasis was immune-independent and targeted the tumor cell compartment, potentially affecting the cancer stem cell. Thus, the integrated outcome of TGFβ antagonism depends on a complex balance between enhancing effective antitumor immunity and disrupting persistent tumor-suppressive effects of TGFβ on the tumor cell. Applying transcriptomic signatures derived from treatment-naïve mouse primary tumors to human breast cancer datasets suggested that patients with breast cancer with high-grade, estrogen receptor-negative disease are most likely to benefit from anti-TGFβ therapy. CONCLUSIONS Contrary to dogma, tumor-suppressive responses to TGFβ are retained in some advanced metastatic tumors. Safe deployment of TGFβ antagonists in the clinic will require good predictive biomarkers.
Collapse
Affiliation(s)
- Yuan Yang
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Howard H Yang
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Binwu Tang
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Alex Man Lai Wu
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Kathleen C Flanders
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Nellie Moshkovich
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Douglas S Weinberg
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michael A Welsh
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jia Weng
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Humberto J Ochoa
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Tiffany Y Hu
- Collaborative Protein Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michelle A Herrmann
- Collaborative Protein Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Jinqiu Chen
- Collaborative Protein Technology Resource, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Elijah F Edmondson
- Pathology Histotechnology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - R Mark Simpson
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Huaitian Liu
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Maxwell P Lee
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Lalage M Wakefield
- Lab of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| |
Collapse
|
9
|
Vitiello GAF, Amarante MK, Banin-Hirata BK, Campos CZ, de Oliveira KB, Losi-Guembarovski R, Watanabe MAE. Authors' reply to the comment "Transforming growth factor beta receptor II (TGFBR2) promoter region polymorphism". Breast Cancer Res Treat 2019; 179:519-520. [PMID: 31562571 DOI: 10.1007/s10549-019-05452-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 09/19/2019] [Indexed: 11/28/2022]
Affiliation(s)
| | - Marla Karine Amarante
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, PR, 86057-970, Brazil
| | - Bruna Karina Banin-Hirata
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, PR, 86057-970, Brazil
| | - Clodoaldo Zago Campos
- Department of Clinical Research, Londrina Cancer Hospital, Londrina, PR, 86015-520, Brazil
| | - Karen Brajão de Oliveira
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, PR, 86057-970, Brazil
| | - Roberta Losi-Guembarovski
- Department of General Biology, Biological Sciences Center, Londrina State University, Londrina, PR, 86057-970, Brazil
| | - Maria Angelica Ehara Watanabe
- Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Londrina, PR, 86057-970, Brazil. .,Laboratory DNA Polymorphisms and Immunology, Department of Pathological Sciences, Biological Sciences Center, Londrina State University, Celso Garcia Cid highway (PR445), Km380, Londrina, PR, 86057-970, Brazil.
| |
Collapse
|
10
|
Transforming growth factor beta receptor II (TGFBR2) promoter region polymorphism in Brazilian breast cancer patients: association with susceptibility, clinicopathological features, and interaction with TGFB1 haplotypes. Breast Cancer Res Treat 2019; 178:207-219. [PMID: 31364002 DOI: 10.1007/s10549-019-05370-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
PURPOSE Transforming growth factor beta (TGFβ) has paradoxical effects in breast cancer (BC), inhibiting initial tumors while promoting aggressive ones. A polymorphism on TGFBR2 promoter region (G-875A, rs3087465) increases TGFβ type II receptor expression and is protective against cancer. Previously, we have shown that TGFB1 variants have subtype-specific roles in BC. This work sought to investigate the association between TGFBR2 and susceptibility and clinicopathological features in BC subgroups. METHODS TGFBR2 G-875A was analyzed through PCR-RFLP in 388 BC patients and 405 neoplasia-free women. Case-control analyses as well as interaction with TGFB1 haplotypes previously associated with BC were tested through age-adjusted logistic regression. Correlations between G-875A and clinicopathological parameters were assessed through Kendall's Tau-b test. All statistical tests were two-tailed (α = 0.05). RESULTS TGFBR2 G-875A was protective against BC in additive, genotypic, and dominant models. In subgroup-stratified analyses, these effects were greater in hormonal receptor-positive and luminal-A tumors, but were not significant in other subgroups. Logistic models including TGFB1 variants showed that in luminal-A tumors, G-875A retained its significance while TGFB1 haplotype showed a trend towards significance; otherwise, in HER2+ tumors TGFB1 variants remained significant while TGFBR2 showed a trend for association. There was no interaction between these genes. In correlation analyses, G-875A positively correlated with histopathological grade in total sample, and a trend towards significance was observed in triple-negative BCs. CONCLUSION These results indicate that G-875A is a protective factor against BC, especially from luminal-A subtype, but may promote anaplasia in established tumors, consistent with TGFβ signaling roles in BC.
Collapse
|
11
|
Scientific reports concerning the impact of interleukin 4, interleukin 10 and transforming growth factor β on cancer cells. Cent Eur J Immunol 2019; 44:190-200. [PMID: 31530989 PMCID: PMC6745546 DOI: 10.5114/ceji.2018.76273] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/12/2018] [Indexed: 02/07/2023] Open
Abstract
Cytokines are signalling proteins generated in most part by immune cells that have critical functions in cellular lifespan. Here we present recent data on three selected anti-inflammatory cytokines: interleukin (IL)-10, IL-4 and transforming growth factor β (TGF-β). IL-10 inhibits the synthesis of major pro-inflammatory cytokines, chemokines, and mediates anti-inflammatory reactions. IL-4 is a multifunctional cytokine which plays a crucial role in the regulation of immune responses and is involved in processes associated with development and differentiation of lymphocytes and regulation of T cell survival. Transforming TGF-β, which in normal cells or pre-cancerous cells, promotes proliferation arrest which represses tumour growth. In this review, we focus on the influence of IL-10, IL-4 and TGF-β on various types of cancer as well as potential of these selected cytokines to serve as new biomarkers which can support effective therapies for cancer patients. This article is presented based on a review of the newest research results.
Collapse
|
12
|
Eltoukhy HS, Sinha G, Moore CA, Gergues M, Rameshwar P. Secretome within the bone marrow microenvironment: A basis for mesenchymal stem cell treatment and role in cancer dormancy. Biochimie 2018; 155:92-103. [PMID: 29859990 DOI: 10.1016/j.biochi.2018.05.018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
The secretome produced by cells within the bone marrow is significant to homeostasis. The bone marrow, a well-studied organ, has multiple niches with distinct roles for supporting stem cell functions. Thus, an understanding of mediators involved in the regulation of stem cells could serve as a model for clinical problems and solutions such as tissue repair and regeneration. The exosome secretome of bone marrow stem cells is a developing area of research with respect to the regenerative potential by bone marrow cell, particularly the mesenchymal stem cells. The bone marrow niche regulates endogenous processes such as hematopoiesis but could also support the survival of tumors such as facilitating the cancer stem cells to exist in dormancy for decades. The bone marrow-derived secretome will be critical to future development of therapeutic strategies for oncologic diseases, in addition to regenerative medicine. This article discusses the importance for parallel studies to determine how the same secretome may compromise safety during the use of stem cells in regenerative medicine.
Collapse
Affiliation(s)
- Hussam S Eltoukhy
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Garima Sinha
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Caitlyn A Moore
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Marina Gergues
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA
| | - Pranela Rameshwar
- Department of Medicine - Division of Hematology/Oncology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
13
|
Liu Z, Kundu-Roy T, Matsuura I, Wang G, Lin Y, Lou YR, Barnard NJ, Wang XF, Huang MT, Suh N, Liu F. Carcinogen 7,12-dimethylbenz[a]anthracene-induced mammary tumorigenesis is accelerated in Smad3 heterozygous mice compared to Smad3 wild type mice. Oncotarget 2018; 7:64878-64885. [PMID: 27588495 PMCID: PMC5323122 DOI: 10.18632/oncotarget.11713] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 08/01/2016] [Indexed: 01/09/2023] Open
Abstract
Previous studies based on cell culture and xenograft animal models suggest that Smad3 has tumor suppressor function for breast cancer during early stages of tumorigenesis. In this report, we show that DMBA (7,12-dimethylbenz[a]anthracene), a chemical carcinogen, induces mammary tumor formation at a significantly higher frequency in the Smad3 heterozygous mice than in the Smad3 wild type mice. This is the first genetic evidence showing that Smad3 inhibits mammary tumor formation in a mouse model. Our findings support the notion that Smad3 has important tumor suppressor function for breast cancer.
Collapse
Affiliation(s)
- Zhengxue Liu
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,College of Life Science & Engineering, Chongqing Three Gorges University, Chongqing, China
| | - Tanima Kundu-Roy
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Isao Matsuura
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.,Division of Molecular Genomics and Medicine, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Guannan Wang
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Yong Lin
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - You-Rong Lou
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Nicola J Barnard
- Department of Pathology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Xiao-Fan Wang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Mou-Tuan Huang
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Nanjoo Suh
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Fang Liu
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| |
Collapse
|
14
|
Velloso FJ, Bianco AFR, Farias JO, Torres NEC, Ferruzo PYM, Anschau V, Jesus-Ferreira HC, Chang THT, Sogayar MC, Zerbini LF, Correa RG. The crossroads of breast cancer progression: insights into the modulation of major signaling pathways. Onco Targets Ther 2017; 10:5491-5524. [PMID: 29200866 PMCID: PMC5701508 DOI: 10.2147/ott.s142154] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cancer is the disease with highest public health impact in developed countries. Particularly, breast cancer has the highest incidence in women worldwide and the fifth highest mortality in the globe, imposing a significant social and economic burden to society. The disease has a complex heterogeneous etiology, being associated with several risk factors that range from lifestyle to age and family history. Breast cancer is usually classified according to the site of tumor occurrence and gene expression profiling. Although mutations in a few key genes, such as BRCA1 and BRCA2, are associated with high breast cancer risk, the large majority of breast cancer cases are related to mutated genes of low penetrance, which are frequently altered in the whole population. Therefore, understanding the molecular basis of breast cancer, including the several deregulated genes and related pathways linked to this pathology, is essential to ensure advances in early tumor detection and prevention. In this review, we outline key cellular pathways whose deregulation has been associated with breast cancer, leading to alterations in cell proliferation, apoptosis, and the delicate hormonal balance of breast tissue cells. Therefore, here we describe some potential breast cancer-related nodes and signaling concepts linked to the disease, which can be positively translated into novel therapeutic approaches and predictive biomarkers.
Collapse
Affiliation(s)
| | | | | | | | | | - Valesca Anschau
- Department of Genetics and Evolutionary Biology, Institute of Biosciences, University of São Paulo, São Paulo, Brazil
| | | | - Ted Hung-Tse Chang
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | | | - Luiz F Zerbini
- Cancer Genomics Group, International Center for Genetic Engineering and Biotechnology (ICGEB), Cape Town, South Africa
| | - Ricardo G Correa
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
15
|
Cantelli G, Crosas-Molist E, Georgouli M, Sanz-Moreno V. TGFΒ-induced transcription in cancer. Semin Cancer Biol 2017; 42:60-69. [PMID: 27586372 PMCID: PMC6137079 DOI: 10.1016/j.semcancer.2016.08.009] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/19/2016] [Indexed: 12/15/2022]
Abstract
The Transforming Growth Factor-beta (TGFβ) pathway mediates a broad spectrum of cellular processes and is involved in several diseases, including cancer. TGFβ has a dual role in tumours, acting as a tumour suppressor in the early phase of tumorigenesis and as a tumour promoter in more advanced stages. In this review, we discuss the effects of TGFβ-driven transcription on all stages of tumour progression, with special focus on lung cancer. Since some TGFβ target genes are specifically involved in promoting metastasis, we speculate that these genes might be good targets to block tumour progression without compromising the tumour suppressor effects of the TGFβ pathway.
Collapse
Affiliation(s)
- Gaia Cantelli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Eva Crosas-Molist
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Mirella Georgouli
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK
| | - Victoria Sanz-Moreno
- Tumour Plasticity Laboratory, Randall Division of Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, London SE1 1UL, UK.
| |
Collapse
|
16
|
Yokobori T, Nishiyama M. TGF-β Signaling in Gastrointestinal Cancers: Progress in Basic and Clinical Research. J Clin Med 2017; 6:jcm6010011. [PMID: 28106769 PMCID: PMC5294964 DOI: 10.3390/jcm6010011] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/31/2016] [Accepted: 01/16/2017] [Indexed: 12/18/2022] Open
Abstract
Transforming growth factor (TGF)-β superfamily proteins have many important biological functions, including regulation of tissue differentiation, cell proliferation, and migration in both normal and cancer cells. Many studies have reported that TGF-β signaling is associated with disease progression and therapeutic resistance in several cancers. Similarly, TGF-β-induced protein (TGFBI)—a downstream component of the TGF-β signaling pathway—has been shown to promote and/or inhibit cancer. Here, we review the state of basic and clinical research on the roles of TGF-β and TGFBI in gastrointestinal cancers.
Collapse
Affiliation(s)
- Takehiko Yokobori
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| | - Masahiko Nishiyama
- Research Program for Omics-based Medical Science, Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
- Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan.
| |
Collapse
|
17
|
Miller LD, Chou JA, Black MA, Print C, Chifman J, Alistar A, Putti T, Zhou X, Bedognetti D, Hendrickx W, Pullikuth A, Rennhack J, Andrechek ER, Demaria S, Wang E, Marincola FM. Immunogenic Subtypes of Breast Cancer Delineated by Gene Classifiers of Immune Responsiveness. Cancer Immunol Res 2016; 4:600-10. [PMID: 27197066 DOI: 10.1158/2326-6066.cir-15-0149] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 02/16/2016] [Indexed: 12/24/2022]
Abstract
The abundance and functional orientation of tumor-infiltrating lymphocytes in breast cancer is associated with distant metastasis-free survival, yet how this association is influenced by tumor phenotypic heterogeneity is poorly understood. Here, a bioinformatics approach defined tumor biologic attributes that influence this association and delineated tumor subtypes that may differ in their ability to sustain durable antitumor immune responses. A large database of breast tumor expression profiles and associated clinical data was compiled, from which the ability of phenotypic markers to significantly influence the prognostic performance of a classification model that incorporates immune cell-specific gene signatures was ascertained. Markers of cell proliferation and intrinsic molecular subtype reproducibly distinguished two breast cancer subtypes that we refer to as immune benefit-enabled (IBE) and immune benefit-disabled (IBD). The IBE tumors, comprised mostly of highly proliferative tumors of the basal-like, HER2-enriched, and luminal B subtypes, could be stratified by the immune classifier into significantly different prognostic groups, while IBD tumors could not, indicating the potential for productive engagement of metastasis-protective immunity in IBE tumors, but not in IBD tumors. The prognostic stratification in IBE was independent of conventional variables. Gene network analysis predicted the activation of TNFα/IFNγ signaling pathways in IBE tumors and the activation of the transforming growth factor-β pathway in IBD tumors. This prediction supports a model in which breast tumors can be distinguished on the basis of their potential for metastasis-protective immune responsiveness. Whether IBE and IBD represent clinically relevant contexts for evaluating sensitivity to immunotherapeutic agents warrants further investigation. Cancer Immunol Res; 4(7); 600-10. ©2016 AACR.
Collapse
Affiliation(s)
- Lance D Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina. The Comprehensive Cancer Center of Wake Forest University, Winston Salem, North Carolina.
| | - Jeff A Chou
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Michael A Black
- Department of Biochemistry, Otago School of Medical Sciences, University of Otago, Dunedin, New Zealand
| | - Cristin Print
- Department of Molecular Medicine and Pathology and Maurice Wilkins Institute, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Julia Chifman
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Angela Alistar
- The Comprehensive Cancer Center of Wake Forest University, Winston Salem, North Carolina. Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Thomas Putti
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, National University Hospital, Singapore
| | - Xiaobo Zhou
- Department of Radiology, Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Davide Bedognetti
- Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Wouter Hendrickx
- Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Ashok Pullikuth
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jonathan Rennhack
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Eran R Andrechek
- Department of Physiology, Michigan State University, East Lansing, Michigan
| | - Sandra Demaria
- Department of Radiation Oncology and Pathology, Weill Cornell Medical College, New York, New York
| | - Ena Wang
- Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - Francesco M Marincola
- Office of the Chief Research Officer, Research Branch, Sidra Medical and Research Center, Doha, Qatar
| |
Collapse
|
18
|
Friberg S, Nyström AM. NANOMEDICINE: will it offer possibilities to overcome multiple drug resistance in cancer? J Nanobiotechnology 2016; 14:17. [PMID: 26955956 PMCID: PMC4784447 DOI: 10.1186/s12951-016-0172-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Accepted: 03/03/2016] [Indexed: 12/12/2022] Open
Abstract
This review is written with the purpose to review the current nanomedicine literature and provide an outlook on the developments in utilizing nanoscale drug constructs in treatment of solid cancers as well as in the potential treatment of multi-drug resistant cancers. No specific design principles for this review have been utilized apart from our active choice to avoid results only based on in vitro studies. Few drugs based on nanotechnology have progressed to clinical trials, since most are based only on in vitro experiments which do not give the necessary data for the research to progress towards pre-clinical studies. The area of nanomedicine has indeed spark much attention and holds promise for improved future therapeutics in the treatment of solid cancers. However, despite much investment few targeted therapeutics have successfully progressed to early clinical trials, indicating yet again that the human body is complicated and that much more understanding of the fundamentals of receptor interactions, physics of nanomedical constructs and their circulation in the body is indeed needed. We believe that nanomedical therapeutics can allow for more efficient treatments of resistant cancers, and may well be a cornerstone for RNA based therapeutics in the future given their general need for shielding from the harsh environment in the blood stream.
Collapse
Affiliation(s)
- Sten Friberg
- Department of Neuroscience, Swedish Medical Nanoscience Center, Karolinska Institutet, Retzius väg 8, 171 77, Stockholm, Sweden.
| | - Andreas M Nyström
- Institute of Environmental Medicine, Karolinska Institutet, Nobels väg 13, 171 77, Stockholm, Sweden.
| |
Collapse
|
19
|
Abstract
AbstractN-cadherin is an adhesion protein, which is important for intercellular interaction. It is involved in cell migration and motility during embryonic development, neuronal synapsis and cancer metastasis. There are several signaling cascades affected by N-cadherin including TGF-β, Rho family. N-cadherin is associated at the cytoplasmic domain with catenins (α, β, γ and p120) to facilitate metastasis. An increase in N-cadherin with down regulation of E-cadherin occurs during epithelial mesenchymal transition. Overexpression of N-cadherin is associated with cell cycle arrest, which correlates with a similar property of cancer stem cells (CSC). Connexin expression, which is important in CSC dormancy, is regulated by N-cadherin. This review discusses the potential of N-cadherin to be involved in maintaining CSCs, and to investigate pathways in N-cadherin expression. A better understanding of the role of N-Cadherin in CSC biology may identify new targets for the treatment of cancer.
Collapse
|
20
|
Bielecka ZF, Czarnecka AM, Szczylik C. Genomic Analysis as the First Step toward Personalized Treatment in Renal Cell Carcinoma. Front Oncol 2014; 4:194. [PMID: 25120953 PMCID: PMC4110478 DOI: 10.3389/fonc.2014.00194] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/09/2014] [Indexed: 12/13/2022] Open
Abstract
Drug resistance mechanisms in renal cell carcinoma (RCC) still remain elusive. Although most patients initially respond to targeted therapy, acquired resistance can still develop eventually. Most of the patients suffer from intrinsic (genetic) resistance as well, suggesting that there is substantial need to broaden our knowledge in the field of RCC genetics. As molecular abnormalities occur for various reasons, ranging from single nucleotide polymorphisms to large chromosomal defects, conducting whole-genome association studies using high-throughput techniques seems inevitable. In principle, data obtained via genome-wide research should be continued and performed on a large scale for the purposes of drug development and identification of biological pathways underlying cancerogenesis. Genetic alterations are mostly unique for each histological RCC subtype. According to recently published data, RCC is a highly heterogeneous tumor. In this paper, the authors discuss the following: (1) current state-of-the-art knowledge on the potential biomarkers of RCC subtypes; (2) significant obstacles encountered in the translational research on RCC; and (3) recent molecular findings that may have a crucial impact on future therapeutic approaches.
Collapse
Affiliation(s)
- Zofia Felicja Bielecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland ; Postgraduate School of Molecular Medicine, Medical University of Warsaw , Warsaw , Poland
| | - Anna Małgorzata Czarnecka
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| | - Cezary Szczylik
- Department of Oncology with the Laboratory of Molecular Oncology, Military Institute of Medicine , Warsaw , Poland
| |
Collapse
|