101
|
Fan C, Qu H, Wang X, Sobhani N, Wang L, Liu S, Xiong W, Zeng Z, Li Y. Cancer/testis antigens: from serology to mRNA cancer vaccine. Semin Cancer Biol 2021; 76:218-231. [PMID: 33910064 DOI: 10.1016/j.semcancer.2021.04.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 02/01/2023]
Abstract
Cancer/testis antigens (CTAs) are a group of tumor antigens expressed in numerous cancer tissues, as well as in the testis and placental tissues. There are over 200 CTAs supported by serology and expression data. The expression patterns of CTAs reflect the similarities between the processes of gametogenesis and tumorigenesis. It is notable that CTAs are highly expressed in three types of cancers (lung cancer, bladder cancer, and skin cancer), all of which have a metal etiology. Here, we review the expression, regulation, and function of CTAs and their translational prospects as cancer biomarkers and treatment targets. Many CTAs are highly immunogenic, tissue-specific, and frequently expressed in cancer tissues but not under physiological conditions, rendering them promising candidates for cancer detection. Some CTAs are associated with clinical outcomes, so they may serve as prognostic biomarkers. A small number of CTAs are membrane-bound, making them ideal targets for chimeric antigen receptor (CAR) T cells. Mounting evidence suggests that CTAs induce humoral or cellular immune responses, providing cancer immunotherapeutic opportunities for T-cell receptors (TCRs), CAR T cell, antibody-based therapy and peptide- or mRNA-based vaccines. Indeed, CTAs are the dominating non-mutated targets in mRNA cancer vaccine development. Clinical trials on CTA TCR and vaccines have shown effectiveness, safety, and tolerance, but these successes are limited to a small number of patients. In-depth studies on CTA expression and function are needed to improve CTA-based immunotherapy.
Collapse
Affiliation(s)
- Chunmei Fan
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China; Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Hongke Qu
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Xu Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Navid Sobhani
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Leiming Wang
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Shuanglin Liu
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan, China.
| | - Yong Li
- Section of Epidemiology and Population Science, Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, United States.
| |
Collapse
|
102
|
Patient-derived organoids as a predictive biomarker for treatment response in cancer patients. NPJ Precis Oncol 2021; 5:30. [PMID: 33846504 PMCID: PMC8042051 DOI: 10.1038/s41698-021-00168-1] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/10/2021] [Indexed: 02/01/2023] Open
Abstract
Effective predictive biomarkers are needed to enable personalized medicine and increase treatment efficacy and survival for cancer patients, thereby reducing toxic side effects and treatment costs. Patient-derived organoids (PDOs) enable individualized tumour response testing. Since 2018, 17 publications have examined PDOs as a potential predictive biomarker in the treatment of cancer patients. We review and provide a pooled analysis of the results regarding the use of PDOs in individualized tumour response testing, focusing on evidence for analytical validity, clinical validity and clinical utility. We identify future perspectives to accelerate the implementation of PDOs as a predictive biomarker in the treatment of cancer patients.
Collapse
|
103
|
Shaw P, Kumar N, Privat-Maldonado A, Smits E, Bogaerts A. Cold Atmospheric Plasma Increases Temozolomide Sensitivity of Three-Dimensional Glioblastoma Spheroids via Oxidative Stress-Mediated DNA Damage. Cancers (Basel) 2021; 13:cancers13081780. [PMID: 33917880 PMCID: PMC8068248 DOI: 10.3390/cancers13081780] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/03/2021] [Accepted: 04/05/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Cold atmospheric plasma (CAP) is gaining increasing interest for cancer treatment, for a wide range of cancer types. The studies performed with CAP as a standalone treatment modality serve as evidence that it can also be a suitable candidate for combination therapy. Temozolomide (TMZ) is used as the gold standard drug for glioblastoma treatment, one of the most aggressive malignant brain tumors in adults that remains incurable despite treatment advances. In this study, we explore whether CAP, a cocktail of reactive oxygen and nitrogen species, can amplify the cytotoxic effect on both TMZ-sensitive and TMZ-resistant glioblastoma multiforme (GBM) in three-dimensional tumor-like tissues through inhibiting the glutathione (GSH)/ glutathione peroxidase 4 (GPX4) antioxidant machinery, which can further lead to DNA damage. Abstract Glioblastoma multiforme (GBM) is the most frequent and aggressive primary malignant brain tumor in adults. Current standard radiotherapy and adjuvant chemotherapy with the alkylating agent temozolomide (TMZ) yield poor clinical outcome. This is due to the stem-like properties of tumor cells and genetic abnormalities in GBM, which contribute to resistance to TMZ and progression. In this study, we used cold atmospheric plasma (CAP) to enhance the sensitivity to TMZ through inhibition of antioxidant signaling (linked to TMZ resistance). We demonstrate that CAP indeed enhances the cytotoxicity of TMZ by targeting the antioxidant specific glutathione (GSH)/glutathione peroxidase 4 (GPX4) signaling. We optimized the threshold concentration of TMZ on five different GBM cell lines (U251, LN18, LN229, U87-MG and T98G). We combined TMZ with CAP and tested it on both TMZ-sensitive (U251, LN18 and LN229) and TMZ-resistant (U87-MG and T98G) cell lines using two-dimensional cell cultures. Subsequently, we used a three-dimensional spheroid model for the U251 (TMZ-sensitive) and U87-MG and T98G (TMZ-resistant) cells. The sensitivity of TMZ was enhanced, i.e., higher cytotoxicity and spheroid shrinkage was obtained when TMZ and CAP were administered together. We attribute the anticancer properties to the release of intracellular reactive oxygen species, through inhibiting the GSH/GPX4 antioxidant machinery, which can lead to DNA damage. Overall, our findings suggest that the combination of CAP with TMZ is a promising combination therapy to enhance the efficacy of TMZ towards the treatment of GBM spheroids.
Collapse
Affiliation(s)
- Priyanka Shaw
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
- Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Antwerp, Belgium
| | - Naresh Kumar
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
- National Institute of Pharmaceutical Education and Research, Guwahati, Guwahati 781125, Assam, India
| | - Angela Privat-Maldonado
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
- Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Antwerp, Belgium
| | - Evelien Smits
- Solid Tumor Immunology Group, Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Antwerp, Belgium
| | - Annemie Bogaerts
- Research Group PLASMANT, Department of Chemistry, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
104
|
Zhu Y, Ferri-Borgogno S, Sheng J, Yeung TL, Burks JK, Cappello P, Jazaeri AA, Kim JH, Han GH, Birrer MJ, Mok SC, Wong STC. SIO: A Spatioimageomics Pipeline to Identify Prognostic Biomarkers Associated with the Ovarian Tumor Microenvironment. Cancers (Basel) 2021; 13:1777. [PMID: 33917869 PMCID: PMC8068305 DOI: 10.3390/cancers13081777] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 01/02/2023] Open
Abstract
Stromal and immune cells in the tumor microenvironment (TME) have been shown to directly affect high-grade serous ovarian cancer (HGSC) malignant phenotypes, however, how these cells interact to influence HGSC patients' survival remains largely unknown. To investigate the cell-cell communication in such a complex TME, we developed a SpatioImageOmics (SIO) pipeline that combines imaging mass cytometry (IMC), location-specific transcriptomics, and deep learning to identify the distribution of various stromal, tumor and immune cells as well as their spatial relationship in TME. The SIO pipeline automatically and accurately segments cells and extracts salient cellular features to identify biomarkers, and multiple nearest-neighbor interactions among tumor, immune, and stromal cells that coordinate to influence overall survival rates in HGSC patients. In addition, SIO integrates IMC data with microdissected tumor and stromal transcriptomes from the same patients to identify novel signaling networks, which would lead to the discovery of novel survival rate-modulating mechanisms in HGSC patients.
Collapse
Affiliation(s)
- Ying Zhu
- Center for Modeling Cancer Development, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA; (Y.Z.); (J.S.)
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Sammy Ferri-Borgogno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.F.-B.); (T.-L.Y.); (A.A.J.)
| | - Jianting Sheng
- Center for Modeling Cancer Development, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA; (Y.Z.); (J.S.)
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| | - Tsz-Lun Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.F.-B.); (T.-L.Y.); (A.A.J.)
| | - Jared K. Burks
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy;
| | - Amir A. Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.F.-B.); (T.-L.Y.); (A.A.J.)
| | - Jae-Hoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.-H.K.); (G.H.H.)
| | - Gwan Hee Han
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul 03722, Korea; (J.-H.K.); (G.H.H.)
| | - Michael J. Birrer
- Winthrop P. Rockefeller Cancer Institute, The University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Samuel C. Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (S.F.-B.); (T.-L.Y.); (A.A.J.)
| | - Stephen T. C. Wong
- Center for Modeling Cancer Development, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA; (Y.Z.); (J.S.)
- Departments of Pathology and Laboratory Medicine and Radiology, Houston Methodist Hospital, Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
105
|
Sung JY, Cheong JH. Pan-Cancer Analysis Reveals Distinct Metabolic Reprogramming in Different Epithelial-Mesenchymal Transition Activity States. Cancers (Basel) 2021; 13:cancers13081778. [PMID: 33917859 PMCID: PMC8068218 DOI: 10.3390/cancers13081778] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Recent genomic classification of tumors has stated that clinically refractory cancers aggregate as a distinct molecular subtype associated with epithelial–mesenchymal transition (EMT). EMT subtype tumors are clinically intractable due to shared malignant characteristics such as poor prognosis and metastasis and are resistant to chemotherapy and immune checkpoint blockades. Therefore, there is an urgent clinical need for the identification of potential therapeutic targets for this tumor subtype. Here, we profiled the metabolic signatures of 9452 samples across 31 cancer types based on EMT activity and identified that ~80 to 90% of cancer types had high carbohydrate and energy metabolism associated with the high EMT state. Furthermore, we identified CHST14 as a potential metabolic target for the EMT subtype for stomach cancer associated with reprogramming of energy metabolism. Our analyses identified metabolic reprogramming associated with EMT, suggesting metabolism-associated targets for clinically refractory cancer subtypes. Abstract Epithelial–mesenchymal transition (EMT) is critical for cancer development, invasion, and metastasis. Its activity influences metabolic reprogramming, tumor aggressiveness, and patient survival. Abnormal tumor metabolism has been identified as a cancer hallmark and is considered a potential therapeutic target. We profiled distinct metabolic signatures by EMT activity using data from 9452 transcriptomes across 31 different cancer types from The Cancer Genome Atlas. Our results demonstrated that ~80 to 90% of cancer types had high carbohydrate and energy metabolism, which were associated with the high EMT group. Notably, among the distinct EMT activities, metabolic reprogramming in different immune microenvironments was correlated with patient prognosis. Nine cancer types showed a significant difference in survival with the presence of high EMT activity. Stomach cancer showed elevated energy metabolism and was associated with an unfavorable prognosis (p < 0.0068) coupled with high expression of CHST14, indicating that it may serve as a potential drug target. Our analyses highlight the prevalence of cancer type-dependent EMT and metabolic reprogramming activities and identified metabolism-associated genes that may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Ji-Yong Sung
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Jae-Ho Cheong
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul 03722, Korea;
- Department of Surgery, Yonsei University College of Medicine, Seoul 03722, Korea
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 03722, Korea
- Correspondence: ; Tel.: +82-2-2228-2094
| |
Collapse
|
106
|
Effect of cell microenvironment on the drug sensitivity of hepatocellular cancer cells. Oncotarget 2021; 12:674-685. [PMID: 33868588 PMCID: PMC8021027 DOI: 10.18632/oncotarget.27910] [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: 09/23/2020] [Accepted: 02/19/2021] [Indexed: 12/28/2022] Open
Abstract
The native hepatocellular cancer (HCC) microenvironment is characterized by more hypoxic, hypoglycemic, and acidic conditions than those used in standard cell culture. This study aimed to investigate whether HCC cells cultured in more native conditions have an altered phenotype and drug sensitivity compared to those cultured in standard conditions. Six HCC cell lines were cultured in “standard” (21% O2, 25 mM glucose) or more “native” (1% O2, 5 mM glucose, 10 mM lactate) conditions. Cells were assessed for growth rates, cell cycle distribution, relevant metabolite and protein levels, genome-wide gene expression, mitochondrial DNA sequence and sensitivity to relevant drugs. Many differences in cellular and molecular phenotypes and drug sensitivity were observed between the cells. HCC cells cultured in native conditions had slower doubling times, increased HK2 and GLUT, lower PHDA and ATP levels, and mutations in mitochondrial DNA. Thirty-one genes, including the hypoxia-associated NDRG1, were differentially expressed between the cells. HCC patients in The Cancer Genome Atlas (TCGA) with tumors with a high score based on these 31 genes had a poorer prognosis than those with a low score (p = 0.002). From 90 comparisons of drug sensitivity, increased resistance and sensitivity for cells cultured in native conditions was observed in 14 (16%) and 8 (9%) comparisons respectively. In conclusion, cells cultured in more native conditions can have a more glycolytic and aggressive phenotype and varied drug sensitivity to those cultured in standard conditions, and may provide new insights to understanding tumor biology and drug development.
Collapse
|
107
|
Stanković T, Ranđelović T, Dragoj M, Stojković Burić S, Fernández L, Ochoa I, Pérez-García VM, Pešić M. In vitro biomimetic models for glioblastoma-a promising tool for drug response studies. Drug Resist Updat 2021; 55:100753. [PMID: 33667959 DOI: 10.1016/j.drup.2021.100753] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
The poor response of glioblastoma to current treatment protocols is a consequence of its intrinsic drug resistance. Resistance to chemotherapy is primarily associated with considerable cellular heterogeneity, and plasticity of glioblastoma cells, alterations in gene expression, presence of specific tumor microenvironment conditions and blood-brain barrier. In an attempt to successfully overcome chemoresistance and better understand the biological behavior of glioblastoma, numerous tri-dimensional (3D) biomimetic models were developed in the past decade. These novel advanced models are able to better recapitulate the spatial organization of glioblastoma in a real time, therefore providing more realistic and reliable evidence to the response of glioblastoma to therapy. Moreover, these models enable the fine-tuning of different tumor microenvironment conditions and facilitate studies on the effects of the tumor microenvironment on glioblastoma chemoresistance. This review outlines current knowledge on the essence of glioblastoma chemoresistance and describes the progress achieved by 3D biomimetic models. Moreover, comprehensive literature assessment regarding the influence of 3D culturing and microenvironment mimicking on glioblastoma gene expression and biological behavior is also provided. The contribution of the blood-brain barrier as well as the blood-tumor barrier to glioblastoma chemoresistance is also reviewed from the perspective of 3D biomimetic models. Finally, the role of mathematical models in predicting 3D glioblastoma behavior and drug response is elaborated. In the future, technological innovations along with mathematical simulations should create reliable 3D biomimetic systems for glioblastoma research that should facilitate the identification and possibly application in preclinical drug testing and precision medicine.
Collapse
Affiliation(s)
- Tijana Stanković
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Teodora Ranđelović
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Miodrag Dragoj
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Sonja Stojković Burić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia
| | - Luis Fernández
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Ignacio Ochoa
- Tissue Microenvironment Lab (TME), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragon 50018, Spain; Centro Investigación Biomédica en Red. Bioingenieria, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Aragon 50018, Spain; Institute for Health Research Aragon (IIS Aragón), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Victor M Pérez-García
- Departamento de Matemáticas, E.T.S.I. Industriales and Instituto de Matemática Aplicada a la Ciencia y la Ingeniería (IMACI), Universidad de Castilla-La Mancha, Ciudad Real, 13071, Spain
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Despota Stefana 142, 11060, Belgrade, Serbia.
| |
Collapse
|
108
|
ADAM28 from both endothelium and gastric cancer cleaves von Willebrand Factor to eliminate von Willebrand Factor-induced apoptosis of gastric cancer cells. Eur J Pharmacol 2021; 898:173994. [PMID: 33675784 DOI: 10.1016/j.ejphar.2021.173994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 02/26/2021] [Accepted: 02/28/2021] [Indexed: 11/20/2022]
Abstract
Disintegrin and metalloproteinase 28 (ADAM28) is a member of the disintegrin and metalloprotease domain (ADAM) family. It is associated with the growth and metastasis of various malignancies in vivo, but its role in gastric cancer remains unclear. The purpose of this study was to investigate the effect of ADAM28 derived from gastric cancer and endothelium on gastric cancer cells and its related mechanisms. In this study, Western blot analysis and q-PCR results showed that ADAM28 was up-regulated in gastric cancer cell lines. The TCGA database showed that patients with high ADAM28 expression had significantly shorter overall survival than those with low ADAM28 expression. By MTT analysis, wound healing assay, and flow cytometry, we found that overexpression/knockdown of ADAM28 expression in gastric cancer cells can regulate cell proliferation, apoptosis and migration in vitro. In addition, overexpression/knockdown of ADAM28 in human umbilical vein endothelial cells (HUVECs) in the upper ventricle can regulate the apoptosis of lower ventricular gastric cancer cells in the co-culture system. Furthermore, ELISA demonstrated that knockdown of ADAM28 from endothelial cells increased the expression of von Willebrand Factor (vWF) in the supernatant. We found that ADAM28 both from gastric cancer cells and HUVECs eliminated vWF-induced apoptosis of gastric cancer cells by cleaving vWF, and the addition of the vWF knockdown plasmid eliminated the increase of integrin β3, p-TP53 and c-Casp3 caused by ADAM28 knockdown. In conclusion, ADAM28 from endothelium and gastric cancer may cleave vWF to eliminate vWF-induced apoptosis of gastric cancer cells and play an pro-metastasis effect.
Collapse
|
109
|
Elmore LW, Greer SF, Daniels EC, Saxe CC, Melner MH, Krawiec GM, Cance WG, Phelps WC. Blueprint for cancer research: Critical gaps and opportunities. CA Cancer J Clin 2021; 71:107-139. [PMID: 33326126 DOI: 10.3322/caac.21652] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
We are experiencing a revolution in cancer. Advances in screening, targeted and immune therapies, big data, computational methodologies, and significant new knowledge of cancer biology are transforming the ways in which we prevent, detect, diagnose, treat, and survive cancer. These advances are enabling durable progress in the goal to achieve personalized cancer care. Despite these gains, more work is needed to develop better tools and strategies to limit cancer as a major health concern. One persistent gap is the inconsistent coordination among researchers and caregivers to implement evidence-based programs that rely on a fuller understanding of the molecular, cellular, and systems biology mechanisms underpinning different types of cancer. Here, the authors integrate conversations with over 90 leading cancer experts to highlight current challenges, encourage a robust and diverse national research portfolio, and capture timely opportunities to advance evidence-based approaches for all patients with cancer and for all communities.
Collapse
Affiliation(s)
- Lynne W Elmore
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - Susanna F Greer
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - Elvan C Daniels
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - Charles C Saxe
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - Michael H Melner
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - Ginger M Krawiec
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - William G Cance
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| | - William C Phelps
- Office of the Chief Medical and Scientific Officer, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
110
|
Wensink E, Bond M, Kucukkose E, May A, Vink G, Koopman M, Kranenburg O, Roodhart J. A review of the sensitivity of metastatic colorectal cancer patients with deficient mismatch repair to standard-of-care chemotherapy and monoclonal antibodies, with recommendations for future research. Cancer Treat Rev 2021; 95:102174. [PMID: 33721596 DOI: 10.1016/j.ctrv.2021.102174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/29/2022]
Abstract
In 5% of metastatic colorectal cancer (mCRC) patients, tumours display a deficient mismatch repair (dMMR) system. Immunotherapy is beneficial in dMMR mCRC patients and has recently been approved by the Food and Drug Administration for patients with unresectable or metastatic dMMR CRC. Although dMMR and proficient MMR (pMMR) CRC tumours are biologically distinct, they are commonly treated with the same chemotherapy and monoclonal antibodies. This includes dMMR mCRC patients who did not respond to immunotherapy (20-30%). However, it is unclear if these treatments are equally beneficial in dMMR mCRC. Of note, dMMR mCRC patients have a worse prognosis compared to pMMR, which may in part be caused by a lower response to treatment. To avoid unnecessary exposure to ineffective treatments and their associated toxicity, it is important to identify which systemic treatments are most beneficial in dMMR mCRC patients, thus improving their outcome. Indeed, future treatment strategies are likely to involve combinations of immunotherapy, chemotherapy and monoclonal antibodies. In this evidence-based review, we summarize clinical trials reporting treatment efficacy of different types of chemotherapy and monoclonal antibodies in dMMR mCRC patients. We also review the biological rationale behind a potential differential benefit of chemotherapy with or without monoclonal antibodies in dMMR mCRC patients. A barrier in the interpretation of preclinical results is the choice of model systems. They largely comprise traditional models, including cell lines and xenografts, rather than more representative models, such as patient-derived organoids. We provide concrete recommendations for clinical investigators and fundamental researchers to accelerate research regarding which systemic therapy is most effective in dMMR mCRC patients.
Collapse
Affiliation(s)
- Emerens Wensink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Marinde Bond
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands; Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584CX Utrecht, the Netherlands
| | - Emre Kucukkose
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Anne May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584CX Utrecht, the Netherlands
| | - Geraldine Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands; Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Godebaldkwartier 419, 3511DT Utrecht, the Netherlands
| | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Onno Kranenburg
- Department of Surgical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands; Utrecht Platform for Organoid Technology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands
| | - Jeanine Roodhart
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584CX Utrecht, the Netherlands.
| |
Collapse
|
111
|
Fontana F, Carollo E, Melling GE, Carter DRF. Extracellular Vesicles: Emerging Modulators of Cancer Drug Resistance. Cancers (Basel) 2021; 13:749. [PMID: 33670185 PMCID: PMC7916933 DOI: 10.3390/cancers13040749] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) have recently emerged as crucial modulators of cancer drug resistance. Indeed, it has been shown that they can directly sequester anti-tumor drugs, decreasing their effective concentration at target sites. Moreover, they facilitate the horizontal transfer of specific bioactive cargoes able to regulate proliferative, apoptotic, and stemness programs in recipient cells, potentially conferring a resistant phenotype to drug-sensitive cancer cells. Finally, EVs can mediate the communication between the tumor and both stromal and immune cells within the microenvironment, promoting treatment escape. In this context, clarifying the EV-driven resistance mechanisms might improve not only tumor diagnosis and prognosis but also therapeutic outcomes. Detailed cellular and molecular events occurring during the development of EV-mediated cancer drug resistance are described in this review article.
Collapse
Affiliation(s)
- Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, via Balzaretti 9, 20133 Milan, Italy
| | - Emanuela Carollo
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK; (E.C.); (G.E.M.)
| | - Genevieve E. Melling
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK; (E.C.); (G.E.M.)
- Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - David R. F. Carter
- Department of Biological and Medical Sciences, Oxford Brookes University, Gipsy Lane, Oxford OX3 0BP, UK; (E.C.); (G.E.M.)
| |
Collapse
|
112
|
Gómez-Oliva R, Domínguez-García S, Carrascal L, Abalos-Martínez J, Pardillo-Díaz R, Verástegui C, Castro C, Nunez-Abades P, Geribaldi-Doldán N. Evolution of Experimental Models in the Study of Glioblastoma: Toward Finding Efficient Treatments. Front Oncol 2021; 10:614295. [PMID: 33585240 PMCID: PMC7878535 DOI: 10.3389/fonc.2020.614295] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/14/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common form of brain tumor characterized by its resistance to conventional therapies, including temozolomide, the most widely used chemotherapeutic agent in the treatment of GBM. Within the tumor, the presence of glioma stem cells (GSC) seems to be the reason for drug resistance. The discovery of GSC has boosted the search for new experimental models to study GBM, which allow the development of new GBM treatments targeting these cells. In here, we describe different strategies currently in use to study GBM. Initial GBM investigations were focused in the development of xenograft assays. Thereafter, techniques advanced to dissociate tumor cells into single-cell suspensions, which generate aggregates referred to as neurospheres, thus facilitating their selective expansion. Concomitantly, the finding of genes involved in the initiation and progression of GBM tumors, led to the generation of mice models for the GBM. The latest advances have been the use of GBM organoids or 3D-bioprinted mini-brains. 3D bio-printing mimics tissue cytoarchitecture by combining different types of cells interacting with each other and with extracellular matrix components. These in vivo models faithfully replicate human diseases in which the effect of new drugs can easily be tested. Based on recent data from human glioblastoma, this review critically evaluates the different experimental models used in the study of GB, including cell cultures, mouse models, brain organoids, and 3D bioprinting focusing in the advantages and disadvantages of each approach to understand the mechanisms involved in the progression and treatment response of this devastating disease.
Collapse
Affiliation(s)
- Ricardo Gómez-Oliva
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Samuel Domínguez-García
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Livia Carrascal
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | | | - Ricardo Pardillo-Díaz
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Cristina Verástegui
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain
| | - Carmen Castro
- Área de Fisiología, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| | - Pedro Nunez-Abades
- Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain.,Departamento de Fisiología, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| | - Noelia Geribaldi-Doldán
- Departamento de Anatomía y Embriología Humanas, Facultad de Medicina, Universidad de Cádiz, Cádiz, Spain.,Instituto de Investigación e Innovación Biomédica de Cádiz (INIBICA), Cádiz, Spain
| |
Collapse
|
113
|
A. Razak SA, Mohd Gazzali A, Fisol FA, M. Abdulbaqi I, Parumasivam T, Mohtar N, A. Wahab H. Advances in Nanocarriers for Effective Delivery of Docetaxel in the Treatment of Lung Cancer: An Overview. Cancers (Basel) 2021; 13:400. [PMID: 33499040 PMCID: PMC7865793 DOI: 10.3390/cancers13030400] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/09/2020] [Accepted: 12/24/2020] [Indexed: 12/24/2022] Open
Abstract
Docetaxel (DCX) is a highly effective chemotherapeutic drug used in the treatment of different types of cancer, including non-small cell lung cancer (NSCLC). The drug is known to have low oral bioavailability due to its low aqueous solubility, poor membrane permeability and susceptibility to hepatic first-pass metabolism. To mitigate these problems, DCX is administered via the intravenous route. Currently, DCX is commercially available as a single vial that contains polysorbate 80 and ethanol to solubilize the poorly soluble drug. However, this formulation causes short- and long-term side effects, including hypersensitivity, febrile neutropenia, fatigue, fluid retention, and peripheral neuropathy. DCX is also a substrate to the drug efflux pump P-glycoprotein (P-gp) that would reduce its concentration within the vicinity of the cells and lead to the development of drug resistance. Hence, the incorporation of DCX into various nanocarrier systems has garnered a significant amount of attention in recent years to overcome these drawbacks. The surfaces of these drug-delivery systems indeed can be functionalized by modification with different ligands for smart targeting towards cancerous cells. This article provides an overview of the latest nanotechnological approaches and the delivery systems that were developed for passive and active delivery of DCX via different routes of administration for the treatment of lung cancer.
Collapse
Affiliation(s)
- S. Aishah A. Razak
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Amirah Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Faisalina Ahmad Fisol
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
- Malaysian Institute of Pharmaceuticals and Nutraceuticals (IPharm), National Institute of Biotechnology Malaysia (NIBM), Ministry of Science, Technology and Innovation (MOSTI), Gelugor, Penang 11700, Malaysia
| | - Ibrahim M. Abdulbaqi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Thaigarajan Parumasivam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Noratiqah Mohtar
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| | - Habibah A. Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden, Penang 11800, Malaysia; (S.A.A.R.); (F.A.F.); (I.M.A.); (T.P.); (N.M.)
| |
Collapse
|
114
|
Helal M, Yan C, Gong Z. Stimulation of hepatocarcinogenesis by activated cholangiocytes via Il17a/f1 pathway in kras transgenic zebrafish model. Sci Rep 2021; 11:1372. [PMID: 33446803 PMCID: PMC7809472 DOI: 10.1038/s41598-020-80621-6] [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: 05/05/2020] [Accepted: 12/11/2020] [Indexed: 01/29/2023] Open
Abstract
It has been well known that tumor progression is dependent on secreted factors not only from tumor cells but also from other surrounding non-tumor cells. In the current study, we investigated the role of cholangiocytes during hepatocarcinogenesis following induction of oncogenic krasV12 expression in hepatocytes using an inducible transgenic zebrafish model. Upon induction of carcinogenesis in hepatocytes, a progressive cell proliferation in cholangiocytes was observed. The proliferative response in cholangiocytes was induced by enhanced lipogenesis and bile acids secretion from hepatocytes through activation of Sphingosine 1 phosphate receptor 2 (S1pr2), a known cholangiocyte receptor involving in cholangiocyte proliferation. Enhancement and inhibition of S1pr2 could accelerate or inhibit cholangiocyte proliferation and hepatocarcinogenesis respectively. Gene expression analysis of hepatocytes and cholangiocytes showed that cholangiocytes stimulated carcinogenesis in hepatocytes via an inflammatory cytokine, Il17a/f1, which activated its receptor (Il17ra1a) on hepatocytes and enhanced hepatocarcinogenesis via an ERK dependent pathway. Thus, the enhancing effect of cholangiocytes on hepatocarcinogenesis is likely via an inflammatory loop.
Collapse
Affiliation(s)
- Mohamed Helal
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, Singapore, Singapore ,grid.419615.e0000 0004 0404 7762Marine Pollution Lab, Marine Environment Division, National Institute of Oceanography and Fisheries, Alexandria, Egypt
| | - Chuan Yan
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Zhiyuan Gong
- grid.4280.e0000 0001 2180 6431Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| |
Collapse
|
115
|
Different Calculation Strategies Are Congruent in Determining Chemotherapy Resistance of Brain Tumors In Vitro. Cells 2020; 9:cells9122689. [PMID: 33333810 PMCID: PMC7765228 DOI: 10.3390/cells9122689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/14/2023] Open
Abstract
In cancer pharmacology, a drug candidate’s therapeutic potential is typically expressed as its ability to suppress cell growth. Different methods in assessing the cell phenotype and calculating the drug effect have been established. However, inconsistencies in drug response outcomes have been reported, and it is still unclear whether and to what extent the choice of data post-processing methods is responsible for that. Studies that systematically examine these questions are rare. Here, we compare three established calculation methods on a collection of nine in vitro models of glioblastoma, exposed to a library of 231 clinical drugs. The therapeutic potential of the drugs is determined on the growth curves, using growth inhibition 50% (GI50) and point-of-departure (PoD) as the criteria. An effect is detected on 36% of the drugs when relying on GI50 and on 27% when using PoD. For the area under the curve (AUC), a threshold of 9.5 or 10 could be set to discriminate between the drugs with and without an effect. GI50, PoD, and AUC are highly correlated. The ranking of substances by different criteria varies somewhat, but the group of the top 20 substances according to one criterion typically includes 17–19 top candidates according to another. In addition to generating preclinical values with high clinical potential, we present off-target appreciation of top substance predictions by interrogating the drug response data of non-cancer cells in our calculation technology.
Collapse
|
116
|
The biomarkers related to immune related adverse events caused by immune checkpoint inhibitors. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:284. [PMID: 33317597 PMCID: PMC7734811 DOI: 10.1186/s13046-020-01749-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023]
Abstract
The enthusiasm for immune checkpoint inhibitors (ICIs), an efficient tumor treatment model different from traditional treatment, is based on their unprecedented antitumor effect, but the occurrence of immune-related adverse events (irAEs) is an obstacle to the prospect of ICI treatment. IrAEs are a discrete toxicity caused by the nonspecific activation of the immune system and can affect almost all tissues and organs. Currently, research on biomarkers mainly focuses on the gastrointestinal tract, endocrine system, skin and lung. Several potential hypotheses concentrate on the overactivation of the immune system, excessive release of inflammatory cytokines, elevated levels of pre-existing autoantibodies, and presence of common antigens between tumors and normal tissues. This review lists the current biomarkers that might predict irAEs and their possible mechanisms for both nonspecific and organ-specific biomarkers. However, the prediction of irAEs remains a major clinical challenge to screen and identify patients who are susceptible to irAEs and likely to benefit from ICIs.
Collapse
|
117
|
Nicoś M, Krawczyk P, Crosetto N, Milanowski J. The Role of Intratumor Heterogeneity in the Response of Metastatic Non-Small Cell Lung Cancer to Immune Checkpoint Inhibitors. Front Oncol 2020; 10:569202. [PMID: 33344229 PMCID: PMC7746867 DOI: 10.3389/fonc.2020.569202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/09/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) represent one of the most promising therapeutic approaches in metastatic non-small cell lung cancer (M-NSCLC). Unfortunately, approximately 50–75% of patients do not respond to this treatment modality. Intratumor heterogeneity (ITH) at the genetic and phenotypic level is considered as a major cause of anticancer therapy failure, including resistance to ICIs. Recent observations suggest that spatial heterogeneity in the composition and spatial organization of the tumor microenvironment plays a major role in the response of M-NSCLC patients to ICIs. In this mini review, we first present a brief overview of the use of ICIs in M-NSCLC. We then discuss the role of genetic and non-genetic ITH on the efficacy of ICIs in patients with M-NSCLC.
Collapse
Affiliation(s)
- Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland.,Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| | - Nicola Crosetto
- Science for Life Laboratory, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
118
|
Singh B, Patwardhan RS, Jayakumar S, Sharma D, Sandur SK. Oxidative stress associated metabolic adaptations regulate radioresistance in human lung cancer cells. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 213:112080. [PMID: 33232882 DOI: 10.1016/j.jphotobiol.2020.112080] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 10/05/2020] [Accepted: 11/07/2020] [Indexed: 01/29/2023]
Abstract
Differential inherent and acquired radioresistance of human lung cancer cells contribute to poor therapeutic outcome and tumor recurrence after radiotherapy. Inherent radioresistance of lung cancer cells is known to be associated with ROSLow cancer stem cells (CSCs). However, mechanism of acquired radioresistance in lung cancer cells is poorly understood. Here, we exposed human lung cancer cells (A549) to a cumulative dose of 40Gy and allowed the radioresistant (RR) survivors to divide and form macroscopic colonies after each fraction of 5Gy dose. The RR subline exhibited enrichment of cytosolic ROSHigh cells without specific increase in mitochondrial ROS levels. We found a concomitant increase in the expression of redox regulatory transcription factor Nrf2 and its dependent antioxidant genes in RR cells and cell cycle delay as compared to parental cells. The treatment of RR cells with Nrf2 inhibitor resulted in decreased clonogenic survival indicating their addiction to Nrf2 for metabolic adaptations under high levels of cytosolic ROS. A causal role of inherent ROS levels in conferring radioresistance was established by sorting ROSHigh and ROSLow populations from parental and RR cells. It was observed that ROSHigh population from both parental and RR cells exhibited radioresistance as observed by clonogenic assay. Interestingly, ROSHigh population of cells exhibited higher levels of cellular thiols in both parental and RR cells. Thus, our observations highlight presence of a novel subpopulation in lung cancer cells, which exhibits radioresistance by maintaining 'oxidative stress' and Nrf2 dependent metabolic adaptations. We also posit Nrf2 pathway as a druggable target for radiosensitization of RR A549 cells.
Collapse
Affiliation(s)
- Babita Singh
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Mumbai 400094, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Sundarraj Jayakumar
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| | - Santosh K Sandur
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India.
| |
Collapse
|
119
|
Craig M, Jenner AL, Namgung B, Lee LP, Goldman A. Engineering in Medicine To Address the Challenge of Cancer Drug Resistance: From Micro- and Nanotechnologies to Computational and Mathematical Modeling. Chem Rev 2020; 121:3352-3389. [PMID: 33152247 DOI: 10.1021/acs.chemrev.0c00356] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance has profoundly limited the success of cancer treatment, driving relapse, metastasis, and mortality. Nearly all anticancer drugs and even novel immunotherapies, which recalibrate the immune system for tumor recognition and destruction, have succumbed to resistance development. Engineers have emerged across mechanical, physical, chemical, mathematical, and biological disciplines to address the challenge of drug resistance using a combination of interdisciplinary tools and skill sets. This review explores the developing, complex, and under-recognized role of engineering in medicine to address the multitude of challenges in cancer drug resistance. Looking through the "lens" of intrinsic, extrinsic, and drug-induced resistance (also referred to as "tolerance"), we will discuss three specific areas where active innovation is driving novel treatment paradigms: (1) nanotechnology, which has revolutionized drug delivery in desmoplastic tissues, harnessing physiochemical characteristics to destroy tumors through photothermal therapy and rationally designed nanostructures to circumvent cancer immunotherapy failures, (2) bioengineered tumor models, which have benefitted from microfluidics and mechanical engineering, creating a paradigm shift in physiologically relevant environments to predict clinical refractoriness and enabling platforms for screening drug combinations to thwart resistance at the individual patient level, and (3) computational and mathematical modeling, which blends in silico simulations with molecular and evolutionary principles to map mutational patterns and model interactions between cells that promote resistance. On the basis that engineering in medicine has resulted in discoveries in resistance biology and successfully translated to clinical strategies that improve outcomes, we suggest the proliferation of multidisciplinary science that embraces engineering.
Collapse
Affiliation(s)
- Morgan Craig
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Bumseok Namgung
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Luke P Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| |
Collapse
|
120
|
Zhao P, Zhang J, Wu A, Zhang M, Zhao Y, Tang Y, Wang B, Chen T, Li F, Zhao Q, Huang Y. Biomimetic codelivery overcomes osimertinib-resistant NSCLC and brain metastasis via macrophage-mediated innate immunity. J Control Release 2020; 329:1249-1261. [PMID: 33129919 DOI: 10.1016/j.jconrel.2020.10.052] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 10/21/2020] [Accepted: 10/26/2020] [Indexed: 12/24/2022]
Abstract
The third-generation of EGFR-TKI osimertinib has been approved as a first-line therapy in NSCLC, representing the most successful advance in molecularly targeted therapy. However, the rapid development of osimertinib resistance renders the unsustainable treatment benefit. Plus, brain metastasis (BMs) is a major mortality cause for NSCLC; there is no drug specifically approved for the osimertinib-resistant BMs of NSCLC yet. To tackle these critical issues, a BBB-permeable biomimetic codelivery system was designed for specifically treating osimertinib-resistant BMs. The T12 peptide-modified albumin nanoparticles coloaded with regorafenib and disulfiram/copper ion chelate repolarized the tumor-promoting CD206hi TGF-β1+ MΦ via inhibition of FROUNT and thus remodeled tumor immune microenvironment. The treatment efficacy in both the subcutaneous H1975/AZDR model and the brain metastasized model demonstrated the effectiveness of the BBB-penetrating combination therapy and the macrophage-mediated innate immunity. This nanotherapeutic combination strategy provides a translational solution to the formidable challenges of overcoming TKI resistance and treating the TKI-resistant BMs.
Collapse
Affiliation(s)
- Pengfei Zhao
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China; State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Jiaxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Aihua Wu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, 826 Zhangheng Rd, Shanghai 201203, China
| | - Meng Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Yuge Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; Nanchang University College of Pharmacy, 461 Bayi Rd, Nanchang 330006, China
| | - Yisi Tang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Bing Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China
| | - Tianxiang Chen
- Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Feng Li
- Auburn University Harrison School of Pharmacy, Auburn, AL 36849, USA
| | - Qiang Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China.
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Rd, Shanghai 201203, China; NMPA Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, Shanghai 201203, China; Zhongshan Branch, The Institute of Drug Research and Development, Chinese Academy of Sciences, Zhongshan 528437, China.
| |
Collapse
|
121
|
Sudan SK, Deshmukh SK, Poosarla T, Holliday NP, Dyess DL, Singh AP, Singh S. Resistin: An inflammatory cytokine with multi-faceted roles in cancer. Biochim Biophys Acta Rev Cancer 2020; 1874:188419. [PMID: 32822824 DOI: 10.1016/j.bbcan.2020.188419] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022]
Abstract
Systemic and organ-confined inflammation has been associated with cancer development and progression. Resistin, initially described as an adipocyte-derived cytokine in mice, is mostly expressed by the macrophages in humans. It has potent pro-inflammatory properties, and its elevated serum levels are detected in cancer patients. Aberrant expression of resistin receptors is also reported in several malignancies and associated with aggressive clinicopathological features. Several lines of evidence demonstrate that resistin, acting through its different receptors, promotes tumor growth, metastasis, and chemoresistance by influencing a variety of cellular phenotypes as well as by modulating the tumor microenvironment. Racially disparate expression of resistin has also attracted much interest, considering prevalent cancer health disparities. This review discusses the aberrant expression of resistin and its receptors, its diverse downstream signaling and impact on tumor growth, metastasis, angiogenesis, and therapy resistance to support its clinical exploitation in biomarker and therapeutic development.
Collapse
Affiliation(s)
- Sarabjeet Kour Sudan
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
| | - Sachin Kumar Deshmukh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Pathology, University of South Alabama, Mobile, AL 36617, USA
| | - Teja Poosarla
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | | | - Donna Lynn Dyess
- Department of Surgery, University of South Alabama, Mobile, AL 36617, USA
| | - Ajay Pratap Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Pathology, University of South Alabama, Mobile, AL 36617, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA
| | - Seema Singh
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA; Department of Pathology, University of South Alabama, Mobile, AL 36617, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL 36688, USA.
| |
Collapse
|
122
|
Hassanzadeh A, Naimi A, Hagh MF, Saraei R, Marofi F, Solali S. Kaempferol Improves TRAIL-Mediated Apoptosis in Leukemia MOLT-4 Cells by the Inhibition of Anti-apoptotic Proteins and Promotion of Death Receptors Expression. Anticancer Agents Med Chem 2020; 19:1835-1845. [PMID: 31364517 DOI: 10.2174/1871520619666190731155859] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL or Apo2L) is a member of the Tumor Necrosis Factor (TNF) superfamily, which stimulates apoptosis in a wide range of cancer cells through binding to Death Receptors 4 and 5 (DR4/5). Nevertheless, TRAIL has noticeable anti-cancer abilities; some cancer cells acquire resistance to TRAIL, and consequently, its potential for inducing apoptosis in target cells is strongly diminished. Acute lymphoblastic leukemia MOLT-4 cell line is one of the most resistant cells to TRAIL that developed resistance to TRAIL through different pathways. TRAIL plus kaempferol was used to eliminate the resistance of the MOLT-4 cells to TRAIL. MATERIALS AND METHODS Firstly, IC50 for kaempferol (95μM) was determined by using the MTT assay. Secondly, the viability of the MOLT-4 cells was assayed by FACS after Annexin V/PI staining, following treatment with TRAIL (50 and 100nM) and kaempferol (95μM) alone and in combination. Finally, the expression levels of the candidate genes involved in resistance to TRAIL were assayed by real-time PCR technique. RESULTS Kaempferol plus TRAIL induced apoptosis robustly in MOLT-4 cells at 12, 24 and 48 hours after treatment. Additionally, it was found that kaempferol could inhibit the expression of c-FLIP, X-IAP, cIAP1/2, FGF-8 and VEGF-beta, and conversely augment the expression of DR4/5 in MOLT-4 cells. CONCLUSION It is suggested that co-treatment of MOLT-4 cells with TRAIL plus kaempferol is a practical and attractive approach to eliminate cancers' resistance to TRAIL by inhibition of the intracellular anti-apoptotic proteins, upregulation of DR4/5 and also by suppression of the VEGF-beta (VEGFB) and FGF-8 expressions.
Collapse
Affiliation(s)
- Ali Hassanzadeh
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Naimi
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid F Hagh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raedeh Saraei
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faroogh Marofi
- Immunology research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
123
|
Nassar ZD, Mah CY, Centenera MM, Irani S, Sadowski MC, Scott JS, Nguyen EV, Nagarajan SR, Moldovan M, Lynn DJ, Daly RJ, Hoy AJ, Butler LM. Fatty Acid Oxidation Is an Adaptive Survival Pathway Induced in Prostate Tumors by HSP90 Inhibition. Mol Cancer Res 2020; 18:1500-1511. [DOI: 10.1158/1541-7786.mcr-20-0570] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
|
124
|
Extracellular vesicle-mediated nucleic acid transfer and reprogramming in the tumor microenvironment. Cancer Lett 2020; 482:33-43. [DOI: 10.1016/j.canlet.2020.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/19/2020] [Accepted: 04/03/2020] [Indexed: 02/06/2023]
|
125
|
Fernandes S, Cassani M, Pagliari S, Filipensky P, Cavalieri F, Forte G. Tumor in 3D: In Vitro Complex Cellular Models to Improve Nanodrugs Cancer Therapy. Curr Med Chem 2020; 27:7234-7255. [PMID: 32586245 DOI: 10.2174/0929867327666200625151134] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/18/2020] [Accepted: 05/31/2020] [Indexed: 02/07/2023]
Abstract
Nanodrugs represent novel solutions to reshuffle repurposed drugs for cancer therapy. They might offer different therapeutic options by combining targeted drug delivery and imaging in unique platforms. Such nanomaterials are deemed to overcome the limitations of currently available treatments, ultimately improving patients' life quality. However, despite these promises being made for over three decades, the poor clinical translation of nanoparticle- based therapies calls for deeper in vit.. and in vivo investigations. Translational issues arise very early during the development of nanodrugs, where complex and more reliable cell models are often replaced by easily accessible and convenient 2D monocultures. This is particularly true in the field of cancer therapy. In fact, 2D monocultures provide poor information about the real impact of the nanodrugs in a complex living organism, especially given the poor mimicry of the solid Tumors Microenvironment (TME). The dense and complex extracellular matrix (ECM) of solid tumors dramatically restricts nanoparticles efficacy, impairing the successful implementation of nanodrugs in medical applications. Herein, we propose a comprehensive guideline of the 3D cell culture models currently available, including their potential and limitations for the evaluation of nanodrugs activity. Advanced culture techniques, more closely resembling the physiological conditions of the TME, might give a better prediction of the reciprocal interactions between cells and nanoparticles and eventually help reconsider the use of old drugs for new applications.
Collapse
Affiliation(s)
- Soraia Fernandes
- International Clinical Research Center (ICRC) of St Anne’s University Hospital, CZ-65691 Brno, Czech Republic
| | - Marco Cassani
- International Clinical Research Center (ICRC) of St Anne’s University Hospital, CZ-65691 Brno, Czech Republic
| | - Stefania Pagliari
- International Clinical Research Center (ICRC) of St Anne’s University Hospital, CZ-65691 Brno, Czech Republic
| | - Petr Filipensky
- St Anne’s University Hospital, CZ-65691 Brno, Czech Republic
| | - Francesca Cavalieri
- School of Science, RMIT University,
Melbourne, VIC, Australia,Dipartimento di Scienze e Tecnologie Chimiche, Università di Roma “Tor
Vergata”, Via Della Ricerca Scientifica, Rome, Italy
| | - Giancarlo Forte
- International Clinical Research Center (ICRC) of St Anne’s University Hospital, CZ-65691 Brno, Czech Republic
| |
Collapse
|
126
|
Implications of the Adiponectin System in Non-Small Cell Lung Cancer Patients: A Case-Control Study. Biomolecules 2020; 10:biom10060926. [PMID: 32570854 PMCID: PMC7356727 DOI: 10.3390/biom10060926] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/05/2020] [Accepted: 06/16/2020] [Indexed: 02/07/2023] Open
Abstract
Alterations of adipose tissue occurring in obesity have been recognized as a major risk factor for several cancers. The relationship between adipose tissue and lung cancer, which is the main cancer-related cause of death worldwide, still requires investigation. Perturbations in the adipokine system are likely to interfere with inter-organ crosstalk in lung cancer, which may influence the lung tumor microenvironment. Adiponectin (Acrp30) expression is deregulated in several cancer types. Acrp30 circulates as oligomers with a Low (LMW), Medium (MMW), and High Molecular Weight (HMW), with the latter mediating the main biological effects. Acrp30 acts through AdipoR1 and AdipoR2 receptors. T-cadherin has been described as a non-signaling receptor. This study's aim was to investigate the regulation of serum Acrp30 and its receptors in sample tissue from non-small cell lung cancer (NSCLC) patients. We recruited 72 NSCLC patients and 60 healthy controls, whom we evaluated in terms of their Acpr30 levels and oligomeric profile. In addition, the expression of AdipoRs in tissues from lung cancer specimens was also measured and compared to coupled healthy lung samples. Our findings show a significant reduction of total Acrp30 levels in NSCLC patients compared to normal subjects, with a specific down-regulation of HMW oligomers. Acrp30 expression was lower in lung adenocarcinoma than other subtypes, regardless of other factors. A significantly higher expression of AdipoR1 was observed, while no differences in R2 and a lower expression of T-cadherin were found in lung cancer specimens compared to normal healthy lung tissues. Involvement of the Acrp30 system in lung cancer may provide new insight into the interaction between adipose tissue and lung and sheds light on its potential ability to influence the lung tumor microenvironment.
Collapse
|
127
|
Ghosh P, Guo Y, Ashrafi A, Chen J, Dey S, Zhong S, Liu J, Campbell J, Konduri PC, Gerberich J, Garrossian M, Mason RP, Zhang L, Liu L. Oxygen-Enhanced Optoacoustic Tomography Reveals the Effectiveness of Targeting Heme and Oxidative Phosphorylation at Normalizing Tumor Vascular Oxygenation. Cancer Res 2020; 80:3542-3555. [PMID: 32546631 DOI: 10.1158/0008-5472.can-19-3247] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 03/20/2020] [Accepted: 06/12/2020] [Indexed: 12/25/2022]
Abstract
Multispectral optoacoustic tomography (MSOT) is an emerging noninvasive imaging modality that can detect real-time dynamic information about the tumor microenvironment in humans and animals. Oxygen enhanced (OE)-MSOT can monitor tumor vasculature and oxygenation during disease development or therapy. Here, we used MSOT and OE-MSOT to examine in mice the response of human non-small cell lung cancer (NSCLC) xenografts to a new class of antitumor drugs, heme-targeting agents heme-sequestering peptide 2 (HSP2) and cyclopamine tartrate (CycT). HSP2 inhibits heme uptake, while CycT inhibits heme synthesis in NSCLC cells, where heme is essential for ATP generation via oxidative phosphorylation. HSP2 and CycT can inhibit ATP generation and thereby suppress NSCLC cell tumorigenic functions. MSOT showed that treatment of NSCLC tumors with HSP2 or CycT reduced total hemoglobin, increased oxygen saturation, and enhanced the amplitude of response to oxygen gas breathing challenge. HSP2 and CycT normalized tumor vasculature and improved tumor oxygenation, where levels of several hypoxia markers in NSCLC tumors were reduced by treatment with HSP2 or CycT. Furthermore, treatment with HSP2 or CycT reduced levels of angiogenic factor VEGFA, its receptor VEGFR1, and vascular marker CD34. Together, our data show that heme-targeting drugs HSP2 and CycT elicit multiple tumor-suppressing functions, such as inhibiting angiogenic function, normalizing tumor vasculature, alleviating tumor hypoxia, and inhibiting oxygen consumption and ATP generation. SIGNIFICANCE: Heme-targeting agents HSP2 and CycT effectively normalize tumor vasculature and alleviate tumor hypoxia, raising the possibility of their combination with chemo-, radio-, and immunotherapies to improve antitumor efficacy.See related commentary by Tomaszewski, p. 3461.
Collapse
Affiliation(s)
- Poorva Ghosh
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Yihang Guo
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Gastrointestinal surgery, The Third XiangYa Hospital of Central South University, Changsha, Hunan, China
| | - Adnin Ashrafi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Jingyu Chen
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas.,Ultrasound Department, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Sanchareeka Dey
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas
| | - Shigen Zhong
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Ultrasound, The General Hospital of Chongqing, Chongqing, China
| | - Jie Liu
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas.,The Third Central Hospital of Tianjin, Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Department of Clinical Laboratory, Hedong District, Tianjin, China
| | - James Campbell
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Jeni Gerberich
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Ralph P Mason
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas.
| | - Li Liu
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
128
|
Wang H, Huang Y. Combination therapy based on nano codelivery for overcoming cancer drug resistance. MEDICINE IN DRUG DISCOVERY 2020. [DOI: 10.1016/j.medidd.2020.100024] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
129
|
Liu H, Ni S, Wang H, Zhang Q, Weng W. Charactering tumor microenvironment reveals stromal-related transcription factors promote tumor carcinogenesis in gastric cancer. Cancer Med 2020; 9:5247-5257. [PMID: 32463580 PMCID: PMC7367614 DOI: 10.1002/cam4.3133] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Transcription factors represent the crucial role of controlling gene transcription in cancer development and progression. However, their functions in gastric cancer have not been thoroughly characterized. For this study, we comprehensively evaluated the correlation between infiltration patterns of tumor microenvironment (TME) cells and TFs expression in the cohort of stomach adenocarcinoma (STAD) from TCGA database. We integrally explored differential expression panel and prognostic value of candidate TFs in TCGA‐STAD cohort. Notably, we found a key transcription factor named HEYL, which its expression level was correlated with stromal component transformation of TME. HEYL was regularly high expressed in gastric cancer and correlated with patients’ poor prognosis. Knockdown of HEYL prominently abrogated the tendency of cell proliferation, migration, and progression in gastric cancer. Consistently, overexpression of HEYL strikingly accelerated the gastric carcinoma development through activating oncogenic signaling pathways and transcriptional activation of cadherin 11 (CDH11). Our findings not only identified the close relationship between TFs and TME phenotype, but also emphasized the crucial importance of TFs, especially HEYL, which could be identified as a candidate biomarker to evaluate prognostic risk and therapeutic effect in gastric cancer.
Collapse
Affiliation(s)
- Haining Liu
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Shujuan Ni
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hanbo Wang
- Jining Medical University, Jining, China
| | - Qiongyan Zhang
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Weiwei Weng
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| |
Collapse
|
130
|
Ravindran Menon D, Hammerlindl H, Torrano J, Schaider H, Fujita M. Epigenetics and metabolism at the crossroads of stress-induced plasticity, stemness and therapeutic resistance in cancer. Theranostics 2020; 10:6261-6277. [PMID: 32483452 PMCID: PMC7255038 DOI: 10.7150/thno.42523] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 02/13/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the recent advances in the treatment of cancers, acquired drug resistance remains a major challenge in cancer management. While earlier studies suggest Darwinian factors driving acquired drug resistance, recent studies point to a more dynamic process involving phenotypic plasticity and tumor heterogeneity in the evolution of acquired drug resistance. Chronic stress after drug treatment induces intrinsic cellular reprogramming and cancer stemness through a slow-cycling persister state, which subsequently drives cancer progression. Both epigenetic and metabolic mechanisms play an important role in this dynamic process. In this review, we discuss how epigenetic and metabolic reprogramming leads to stress-induced phenotypic plasticity and acquired drug resistance, and how the two reprogramming mechanisms crosstalk with each other.
Collapse
|
131
|
Han S, Kim S, Chen Z, Shin HK, Lee SY, Moon HE, Paek SH, Park S. 3D Bioprinted Vascularized Tumour for Drug Testing. Int J Mol Sci 2020; 21:E2993. [PMID: 32340319 PMCID: PMC7215771 DOI: 10.3390/ijms21082993] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/16/2020] [Accepted: 04/21/2020] [Indexed: 12/29/2022] Open
Abstract
An in vitro screening system for anti-cancer drugs cannot exactly reflect the efficacy of drugs in vivo, without mimicking the tumour microenvironment (TME), which comprises cancer cells interacting with blood vessels and fibroblasts. Additionally, the tumour size should be controlled to obtain reliable and quantitative drug responses. Herein, we report a bioprinting method for recapitulating the TME with a controllable spheroid size. The TME was constructed by printing a blood vessel layer consisting of fibroblasts and endothelial cells in gelatine, alginate, and fibrinogen, followed by seeding multicellular tumour spheroids (MCTSs) of glioblastoma cells (U87 MG) onto the blood vessel layer. Under MCTSs, sprouts of blood vessels were generated and surrounding MCTSs thereby increasing the spheroid size. The combined treatment involving the anti-cancer drug temozolomide (TMZ) and the angiogenic inhibitor sunitinib was more effective than TMZ alone for MCTSs surrounded by blood vessels, which indicates the feasibility of the TME for in vitro testing of drug efficacy. These results suggest that the bioprinted vascularized tumour is highly useful for understanding tumour biology, as well as for in vitro drug testing.
Collapse
Affiliation(s)
- Seokgyu Han
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (S.H.); (Z.C.)
| | - Sein Kim
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Zhenzhong Chen
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (S.H.); (Z.C.)
| | - Hwa Kyoung Shin
- Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan 50612, Korea;
- Korean Medical Science Research Center for Healthy-Aging, Pusan National University, Yangsan 50612, Korea
- Graduate Training Program of Korean Medicine for Healthy-Aging, Pusan National University, Yangsan 50612, Korea
| | - Seo-Yeon Lee
- Department of Pharmacology, School of Medicine, Wonkwang University, Iksan 54538, Korea;
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (H.E.M.); (S.H.P.)
- Cancer Research Institute, Hypoxia Ischemia Disease Institute, Seoul National University, Seoul 03080, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University Hospital, Seoul 03080, Korea; (H.E.M.); (S.H.P.)
- Cancer Research Institute, Hypoxia Ischemia Disease Institute, Seoul National University, Seoul 03080, Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, Korea; (S.H.); (Z.C.)
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, Korea;
- Institute of Quantum Biophysics (iQB), Sungkyunkwan University, Suwon 16419, Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
132
|
Fong EJ, Strelez C, Mumenthaler SM. A Perspective on Expanding Our Understanding of Cancer Treatments by Integrating Approaches from the Biological and Physical Sciences. SLAS DISCOVERY 2020; 25:672-683. [PMID: 32297829 PMCID: PMC7372587 DOI: 10.1177/2472555220915830] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Multicellular systems such as cancer suffer from immense complexity. It is imperative to capture the heterogeneity of these systems across scales to achieve a deeper understanding of the underlying biology and develop effective treatment strategies. In this perspective article, we will discuss how recent technologies and approaches from the biological and physical sciences have transformed traditional ways of measuring, interpreting, and treating cancer. During the SLAS 2019 Annual Meeting, SBI2 hosted a Special Interest Group (SIG) on this topic. Academic and industry leaders engaged in discussions surrounding what biological model systems are appropriate to study cancer complexity, what assays are necessary to interrogate this complexity, and how physical sciences approaches may be useful to detangle this complexity. In particular, we examined the utility of mathematical models in predicting cancer progression and treatment response when tightly integrated with reproducible, quantitative, and dynamic biological measurements achieved using high-content imaging and analysis. The dialogue centered around the impetus for convergent biosciences, bringing new perspectives to cancer research to further understand this complex adaptive system and successfully intervene therapeutically.
Collapse
Affiliation(s)
- Emma J Fong
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Carly Strelez
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
133
|
Kato M, Onoyama I, Yoshida S, Cui L, Kawamura K, Kodama K, Hori E, Matsumura Y, Yagi H, Asanoma K, Yahata H, Itakura A, Takeda S, Kato K. Dual-specificity phosphatase 6 plays a critical role in the maintenance of a cancer stem-like cell phenotype in human endometrial cancer. Int J Cancer 2020; 147:1987-1999. [PMID: 32159851 PMCID: PMC7496376 DOI: 10.1002/ijc.32965] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/12/2020] [Accepted: 03/04/2020] [Indexed: 12/16/2022]
Abstract
The prognosis of patients with high‐grade or advanced‐stage endometrial cancer remains poor. As cancer stem‐like cells (CSCs) are thought to be associated with endometrial cancers, it is essential to investigate the molecular mechanisms that regulate endometrial CSCs. Dual‐specificity phosphatase 6 (DUSP6) functions as a negative‐feedback regulator of MAPK–ERK1/2 signaling, but its role in endometrial cancer remains unknown. We investigated whether DUSP6 is involved in cancer cell stemness using endometrial cancer cell lines and specimens from endometrial cancer patients. DUSP6 induced the expression of CSC‐related genes including ALDH1, Nanog, SOX2 and Oct4A, increased the population of cells in the G0/G1 phase, and promoted sphere formation ability. DUSP6 knockdown resulted in reduced cell invasion and metastasis, whereas DUSP6 overexpression inhibited apoptosis under serum‐free conditions. Moreover, DUSP6 decreased phosphorylated ERK1/2 and increased phosphorylated Akt levels, which potentially induces CSC features. In patients with endometrial cancers, DUSP6 expression was determined using immunohistochemistry, and based on the results, the patients were dichotomized into high‐ and low‐DUSP6‐expression groups. Progression‐free survival and overall survival were significantly shorter in the high‐DUSP6‐expression group. These results suggest that DUSP6 has potential value as a biomarker of CSCs and as a target of therapies designed to eliminate CSCs in endometrial cancer. What's new? Although cancer stem‐like cells (CSCs) are involved in human endometrial cancers, the underlying molecular mechanisms and biomarkers for CSCs in endometrial cancers remain elusive. Here, the authors found that DUSP6 plays an important role in regulating endometrial CSC phenotypes by increasing self‐renewal ability and starvation resistance. DUSP6 expression was required for inducing invasion and metastasis and resulted in ERK1/2 dephosphorylation and Akt phosphorylation, which potentially contribute to the promotion of CSC phenotypes. As DUSP6 expression was also positively associated with worse progression‐free and overall survival, DUSP6 represents a potential biomarker for endometrial CSCs and a therapeutic target in endometrial cancers.
Collapse
Affiliation(s)
- Masaya Kato
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
- Department of Obstetrics and GynecologySchool of Medical Sciences, Juntendo UniversityTokyoJapan
| | - Ichiro Onoyama
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Sachiko Yoshida
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Lin Cui
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Keiko Kawamura
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Keisuke Kodama
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Emiko Hori
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Yumiko Matsumura
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Hiroshi Yagi
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Kazuo Asanoma
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Hideaki Yahata
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Atsuo Itakura
- Department of Obstetrics and GynecologySchool of Medical Sciences, Juntendo UniversityTokyoJapan
| | - Satoru Takeda
- Department of Obstetrics and GynecologySchool of Medical Sciences, Juntendo UniversityTokyoJapan
| | - Kiyoko Kato
- Department of Obstetrics and GynecologySchool of Medical Sciences, Kyushu UniversityFukuokaJapan
| |
Collapse
|
134
|
Kabekkodu SP, Shukla V, Varghese VK, Adiga D, Vethil Jishnu P, Chakrabarty S, Satyamoorthy K. Cluster miRNAs and cancer: Diagnostic, prognostic and therapeutic opportunities. WILEY INTERDISCIPLINARY REVIEWS. RNA 2020; 11:e1563. [PMID: 31436881 DOI: 10.1002/wrna.1563] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/05/2019] [Accepted: 07/25/2019] [Indexed: 02/06/2023]
Abstract
MiRNAs are class of noncoding RNA important for gene expression regulation in many plants, animals and viruses. MiRNA clusters contain a set of two or more miRNA encoding genes, transcribed together as polycistronic miRNAs. Currently, there are approximately 159 miRNA clusters reported in the human genome consisting of miRNAs ranging from two or more miRNA genes. A large proportion of clustered miRNAs resides in and around the fragile sites or cancer associated genomic hotspots and plays an important role in carcinogenesis. Altered expression of miRNA cluster can be pro-tumorigenic or anti-tumorigenic and can be targeted for clinical management of cancer. Over the past few years, manipulation of miRNA clusters expression is attempted for experimental purpose as well as for diagnostic, prognostic and therapeutic applications in cancer. Re-expression of miRNAs by epigenetic therapy, genome editing such as clustered regulatory interspaced short palindromic repeats (CRISPR) and miRNA mowers showed promising results in cancer therapy. In this review, we focused on the potential of miRNA clusters as a biomarker for diagnosis, prognosis, targeted therapy as well as strategies for modulating their expression in a therapeutic context. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA Processing > Processing of Small RNAs RNA in Disease and Development > RNA in Disease Regulatory RNAs/RNAi/Riboswitches > Biogenesis of Effector Small RNAs.
Collapse
Affiliation(s)
- Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vaibhav Shukla
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Vinay Koshy Varghese
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Padacherri Vethil Jishnu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sanjiban Chakrabarty
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
135
|
Li F, Zhao S, Cui Y, Guo T, Qiang J, Xie Q, Yu W, Guo W, Deng W, Gu C, Wu T. α1,6-Fucosyltransferase (FUT8) regulates the cancer-promoting capacity of cancer-associated fibroblasts (CAFs) by modifying EGFR core fucosylation (CF) in non-small cell lung cancer (NSCLC). Am J Cancer Res 2020; 10:816-837. [PMID: 32266093 PMCID: PMC7136908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 02/28/2020] [Indexed: 06/11/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are the main cancer-promoting component in the tumor microenvironment (TME) of non-small cell lung cancer (NSCLC). α1,6-Fucosyltransferase (FUT8), the key enzyme catalyzing core α1,6-fucosylation (CF), plays a promoting role in multiple malignancies. In the current study, we investigated the function of FUT8 in CAFs and elucidated the mechanism through which FUT8 regulates the cancer-promoting capacity of CAFs in NSCLC. A bioinformatics analysis was performed to reveal the relationship between FUT8 and CAFs. Resected specimens from NSCLC patients were analyzed to assess the expression of FUT8 in CAFs. Primary CAFs and normal lung fibroblasts (NLFs) were extracted from NSCLC patient specimens and were co-cultured with NSCLC cell lines in a novel 3D-printed non-contact co-culture device. An In vivo CAF/NSCLC co-injection tumorigenesis assay was performed using nude mice to study the function of FUT8/CF in TME formation. The current study revealed that FUT8-mediated CF in CAFs plays a positive role in the cancer-promoting capacity of these cells. FUT8 overexpression was observed in CAFs isolated from some lung adenocarcinoma cases. Further investigation showed that FUT8/CF in CAFs promoted the formation of an invasive and malignant TME in vivo and in vitro, and the resulting NSCLC cells exhibited faster proliferation and increased invasiveness. EGFR signaling exerts a catalytic effect on the cancer-promoting capacity of CAFs and is regulated by the CF modification of the EGFR protein.
Collapse
Affiliation(s)
- Fengzhou Li
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Shilei Zhao
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Yanwei Cui
- Zhongshan Hospital, Dalian UniversityDalian, China
| | - Tao Guo
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Jiaqi Qiang
- Dalian Municipal Central Hospital Affiliated to Dalian Medical UniversityDalian, China
| | - Qiang Xie
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cell, Dalian Medical UniversityDalian, China
| | - Wei Guo
- Institute of Cancer Stem Cell, Dalian Medical UniversityDalian, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Lab of Oncology in South ChinaGuangzhou, China
| | - Chundong Gu
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Taihua Wu
- The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| |
Collapse
|
136
|
Miyazoe Y, Miuma S, Miyaaki H, Kanda Y, Nakashiki S, Sasaki R, Haraguchi M, Shibata H, Honda T, Taura N, Nakao K. Extracellular vesicles from senescent hepatic stellate cells promote cell viability of hepatoma cells through increasing EGF secretion from differentiated THP-1 cells. Biomed Rep 2020; 12:163-170. [PMID: 32190304 PMCID: PMC7054706 DOI: 10.3892/br.2020.1279] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 01/17/2020] [Indexed: 12/28/2022] Open
Abstract
Since the discovery of the senescence-associated secretory phenotype, the role of senescent hepatic stellate cells (HSCs) in hepatocellular carcinoma (HCC) development has gained increasing attention. Similar to cytokines, extracellular vesicles (EVs) are essential for intercellular communication. However, the function of EVs derived from senescent HSCs in HCC progression has not been extensively studied. The aims of the present study were to characterize the EVs derived from senescent HSCs and determine their role in the tumor microenvironment. Cellular senescence was induced in human hepatic stellate cells (HHSteCs) with various concentrations of etoposide. Induction was confirmed using EdU staining and 53BP1 and p21 immunostaining. EVs were isolated by ultracentrifugation and analyzed by nanoparticle tracking analysis. Multiplex immunoassays were used to compare the levels of growth factors secreted from hepatoma cell lines and macrophage cells pretreated with EVs derived from senescent HHSteCs (senescent EVs) with those pretreated with EVs derived from normal cultured HHSteCs (normal EVs). Treatment with 25 µM etoposide for 3 days was the most effective at inducing senescence in HHSteCs. This finding was confirmed by induction of irreversible cell-cycle arrest, upregulation of 53BP1 and p21 expression, and increased SA-β-gal staining. Senescent HHSteCs released increased quantities of EV particles compared with normally cultured HHSteCs. Multiplex analysis revealed that there was no difference between hepatoma cell lines treated with normal EVs and those treated with senescent EVs in growth factor secretion. In contrast, the secretion of epidermal growth factor (EGF) was increased by macrophage cells treated with senescent EVs compared with those treated with normal EVs. Furthermore, senescent EVs did not affect the viability of hepatoma cells but increased the viability of hepatoma cells co-cultured with macrophage cells. In conclusion, the release of EVs from senescent HSCs was higher compared with normal HSCs. Furthermore, senescent EVs promoted HCC development by upregulating EGF secretion from macrophages.
Collapse
Affiliation(s)
- Yuri Miyazoe
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Satoshi Miuma
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hisamitsu Miyaaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yasuko Kanda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Suguru Nakashiki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ryu Sasaki
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Masafumi Haraguchi
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Hidetaka Shibata
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Naota Taura
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kazuhiko Nakao
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| |
Collapse
|
137
|
Bokas A, Papakotoulas P, Sarantis P, Papadimitropoulou A, Papavassiliou AG, Karamouzis MV. Mechanisms of the Antitumor Activity of Low Molecular Weight Heparins in Pancreatic Adenocarcinomas. Cancers (Basel) 2020; 12:432. [PMID: 32069809 PMCID: PMC7072375 DOI: 10.3390/cancers12020432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/01/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer treatment in the last decade. Despite the progress in immunotherapy, most pancreatic cancer patients still do not derive benefit when receiving immune-based therapies. Recently, resistance mechanisms to immune therapies have been mainly focused on tumor microenvironment properties. Pancreatic cancer is considered one of the most lethal and difficult to treat tumors due to its highly immunosuppressive and desmoplastic microenvironment. Low molecular weight heparins (LMWHs) have been used for the treatment and prevention of thromboembolic disease in these patients. However, many nonanticoagulant properties attributed to LMWHs have been described. Exploiting LMWH properties in a combined treatment modality with immune checkpoint inhibition and chemotherapy could provide a new approach in the management of pancreatic adenocarcinoma patients. The ability of LMWH to interfere with various aspects of the tumor microenvironment could result in both the alleviation of immunosuppression and improvement in drug delivery within the tumor, leading to higher cancer cell destruction rates and more potent immune system activity that would, ultimately, lead to better patient outcomes.
Collapse
Affiliation(s)
- Alexandros Bokas
- 1st Department of Medical Oncology, Theagenion Hospital, 54007 Thessaloniki, Greece; (A.B.); (P.P.)
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Pavlos Papakotoulas
- 1st Department of Medical Oncology, Theagenion Hospital, 54007 Thessaloniki, Greece; (A.B.); (P.P.)
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Adriana Papadimitropoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
- First Department of Internal Medicine, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
138
|
Uhlig S, Wuhrer A, Berlit S, Tuschy B, Sütterlin M, Bieback K. Intraoperative radiotherapy for breast cancer treatment efficiently targets the tumor bed preventing breast adipose stromal cell outgrowth. Strahlenther Onkol 2020; 196:398-404. [PMID: 32030446 PMCID: PMC7089893 DOI: 10.1007/s00066-020-01586-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/16/2020] [Indexed: 01/17/2023]
Abstract
OBJECTIVES Mesenchymal stromal cells (MSC) in bone marrow have been shown to be radioresistant, which is related to pronounced DNA repair mechanisms. Intraoperative radiotherapy (IORT) during breast-conserving surgery for early breast cancer is an innovative technique applying low energy x‑ray to the tumor bed immediately after removal of the tumor. IORT is considered to reduce the risk of local tumor recurrence by directly targeting cells of the tumor bed and altering the local microenvironment. Aim of this study was to investigate whether IORT affects the outgrowth potential of breast adipose tissue-derived MSC (bASC) as part of the tumor bed. MATERIALS AND METHODS After surgical tumor resection, biopsies of the tumor bed were taken before (pre IORT) and after IORT (post IORT) and processed applying well-established protocols for ASC isolation and characterization. RESULTS In all, 95% of pre IORT tumor bed samples yielded persistently outgrowing bASC with typical ASC characteristics: fibroblastoid morphology, proliferation, adipogenic and osteogenic differentiation and ASC surface marker expression. However, none of the post IORT samples yielded persistent outgrowth of bASC. CONCLUSIONS After breast-conserving surgery, approximately 90% of local recurrences emerge in close proximity to the initial tumor bed, potentially reflecting a significant contribution of the tumor bed to relapse. Our data show that IORT, besides the proven effect on breast cancer cells, efficiently modifies the tumor environment by having an impact on tumor bed bASC. This effect on tumor bed stromal cells might contribute to reduce the risk of tumor relapse and metastases.
Collapse
Affiliation(s)
- Stefanie Uhlig
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, German Red Cross Blood Donor Services, Heidelberg University, Friedrich-Ebert Str. 107, 68167, Mannheim, Germany
- FlowCore Mannheim, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl Str. 13-17, 68167, Mannheim, Germany
| | - Anne Wuhrer
- Department of Obstetrics and Gynecology, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Sebastian Berlit
- Department of Obstetrics and Gynecology, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Benjamin Tuschy
- Department of Obstetrics and Gynecology, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Marc Sütterlin
- Department of Obstetrics and Gynecology, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, German Red Cross Blood Donor Services, Heidelberg University, Friedrich-Ebert Str. 107, 68167, Mannheim, Germany.
- FlowCore Mannheim, Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
139
|
Karsch-Bluman A, Benny O. Necrosis in the Tumor Microenvironment and Its Role in Cancer Recurrence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:89-98. [PMID: 32030649 DOI: 10.1007/978-3-030-35727-6_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer recurrence is one of the most imminent problems in the current world of medicine, and it is responsible for most of the cancer-related death rates worldwide. Long-term administration of anticancer cytotoxic drugs may act as a double-edged sword, as necrosis may lead to renewed cancer progression and treatment resistance. The lack of nutrients, coupled with the induced hypoxia, triggers cell death and secretion of signals that affect the tumor niche. Many efforts have been made to better understand the contribution of hypoxia and metabolic stress to cancer progression and resistance, but mostly with respect to inflammation. Here we provide an overview of the direct anticancer effects of necrotic signals, which are not necessarily mediated by inflammation and the role of DAMPs (damage-associated molecular patterns) on the formation of a pro-cancerous environment.
Collapse
Affiliation(s)
- Adi Karsch-Bluman
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofra Benny
- The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
140
|
Yaman S, Chintapula U, Rodriguez E, Ramachandramoorthy H, Nguyen KT. Cell-mediated and cell membrane-coated nanoparticles for drug delivery and cancer therapy. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:879-911. [PMID: 33796822 PMCID: PMC8011581 DOI: 10.20517/cdr.2020.55] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
Nanotechnology-based drug delivery platforms have been developed over the last two decades because of their favorable features in terms of improved drug bioavailability and stability. Despite recent advancement in nanotechnology platforms, this approach still falls short to meet the complexity of biological systems and diseases, such as avoiding systemic side effects, manipulating biological interactions and overcoming drug resistance, which hinders the therapeutic outcomes of the NP-based drug delivery systems. To address these issues, various strategies have been developed including the use of engineered cells and/or cell membrane-coated nanocarriers. Cell membrane receptor profiles and characteristics are vital in performing therapeutic functions, targeting, and homing of either engineered cells or cell membrane-coated nanocarriers to the sites of interest. In this context, we comprehensively discuss various cell- and cell membrane-based drug delivery approaches towards cancer therapy, the therapeutic potential of these strategies, and the limitations associated with engineered cells as drug carriers and cell membrane-associated drug nanocarriers. Finally, we review various cell types and cell membrane receptors for their potential in targeting, immunomodulation and overcoming drug resistance in cancer.
Collapse
Affiliation(s)
- Serkan Yaman
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Yaman S and Chintapula U contributed equally to this work
| | - Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Yaman S and Chintapula U contributed equally to this work
| | - Edgar Rodriguez
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Harish Ramachandramoorthy
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
- Correspondence Address: Dr. Kytai T. Nguyen, Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd ERB244, Arlington, TX 76010, USA. E-mail:
| |
Collapse
|
141
|
Nunes SC. Exploiting Cancer Cells Metabolic Adaptability to Enhance Therapy Response in Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:297-310. [PMID: 32130705 DOI: 10.1007/978-3-030-34025-4_15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite all the progresses developed in prevention and new treatment approaches, cancer is the second leading cause of death worldwide, being chemoresistance a pivotal barrier in cancer management. Cancer cells present several mechanisms of drug resistance/tolerance and recently, growing evidence have been supporting a role of metabolism reprograming per se as a driver of chemoresistance. In fact, cancer cells display several adaptive mechanisms that allow the emergency of chemoresistance, revealing cancer as a disease that adapts and evolve along with the treatment. Therefore, clinical protocols that take into account the adaptive potential of cancer cells should be more effective than the current traditional standard protocols on the fighting against cancer.In here, some of the recent findings on the role of metabolism reprograming in cancer chemoresistance emergence will be discussed, as the potential evolutionary strategies that could unable these adaptations, hence allowing to prevent the emergency of treatment resistance, changing cancer outcome.
Collapse
Affiliation(s)
- Sofia C Nunes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| |
Collapse
|
142
|
Nunes SC. Tumor Microenvironment - Selective Pressures Boosting Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:35-49. [PMID: 32130692 DOI: 10.1007/978-3-030-34025-4_2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In 2018, 9.6 million deaths from cancer were estimated, being this disease the second leading cause of death worldwide. Notwithstanding all the efforts developed in prevention, diagnosis and new treatment approaches, chemoresistance seems to be inevitable, leading to cancer progression, recurrence and affecting the outcome of the disease. As more and more evidence support that cancer is an evolutionary and ecological process, this concept is rarely applied in the clinical context. In fact, cancer cells emerge and progress within an ecological niche - the tumor microenvironment - that is shared with several other cell types and that is continuously changing. Therefore, the tumor microenvironment imposes several selective pressures on cancer cells such as acidosis, hypoxia, competition for space and resources, immune predation and anti-cancer therapies, that cancer cells must be able to adapt to or will face extinction.In here, the role of the tumor microenvironment selective pressures on cancer progression will be discussed, as well as the targeting of its features/components as strategies to fight cancer.
Collapse
Affiliation(s)
- Sofia C Nunes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| |
Collapse
|
143
|
Carreira-Barbosa F, Nunes SC. Wnt Signaling: Paths for Cancer Progression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1219:189-202. [PMID: 32130700 DOI: 10.1007/978-3-030-34025-4_10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Wnt signaling pathways are well known for having several pivotal roles during embryonic development. However, the same developmental signaling pathways also present key roles in cancer initiation and progression. In this chapter, several issues regarding the roles of both canonical and non-canonical Wnt signaling pathways in cancer will be explored, mainly concerning their role in the maintenance of cancer stemness, in the metabolism reprograming of cancer cells and in the modulation of the tumor microenvironment. The role of Wnt signaling cascades in the response of cancer cells to anti-cancer treatments will be also discussed, as well as its potential therapeutic targeting during cancer treatment. Collectively, increasing evidence has been supporting pivotal roles of Wnt signaling in several features of cancer biology, however; a lot is still to be elucidated.
Collapse
Affiliation(s)
| | - Sofia C Nunes
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School | Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Lisbon, Portugal
| |
Collapse
|
144
|
Huang EW, Bhope A, Lim J, Sinha S, Emad A. Tissue-guided LASSO for prediction of clinical drug response using preclinical samples. PLoS Comput Biol 2020; 16:e1007607. [PMID: 31967990 PMCID: PMC6975549 DOI: 10.1371/journal.pcbi.1007607] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 12/15/2019] [Indexed: 12/12/2022] Open
Abstract
Prediction of clinical drug response (CDR) of cancer patients, based on their clinical and molecular profiles obtained prior to administration of the drug, can play a significant role in individualized medicine. Machine learning models have the potential to address this issue but training them requires data from a large number of patients treated with each drug, limiting their feasibility. While large databases of drug response and molecular profiles of preclinical in-vitro cancer cell lines (CCLs) exist for many drugs, it is unclear whether preclinical samples can be used to predict CDR of real patients. We designed a systematic approach to evaluate how well different algorithms, trained on gene expression and drug response of CCLs, can predict CDR of patients. Using data from two large databases, we evaluated various linear and non-linear algorithms, some of which utilized information on gene interactions. Then, we developed a new algorithm called TG-LASSO that explicitly integrates information on samples' tissue of origin with gene expression profiles to improve prediction performance. Our results showed that regularized regression methods provide better prediction performance. However, including the network information or common methods of including information on the tissue of origin did not improve the results. On the other hand, TG-LASSO improved the predictions and distinguished resistant and sensitive patients for 7 out of 13 drugs. Additionally, TG-LASSO identified genes associated with the drug response, including known targets and pathways involved in the drugs' mechanism of action. Moreover, genes identified by TG-LASSO for multiple drugs in a tissue were associated with patient survival. In summary, our analysis suggests that preclinical samples can be used to predict CDR of patients and identify biomarkers of drug sensitivity and survival.
Collapse
Affiliation(s)
- Edward W. Huang
- Department of Computer Science, University of Illinois at Urbana-Champaign, Illinois, United States of America
| | - Ameya Bhope
- Department of Electrical and Computer Engineering, McGill University, Canada
| | - Jing Lim
- Department of Computer Science, University of Illinois at Urbana-Champaign, Illinois, United States of America
| | - Saurabh Sinha
- Department of Computer Science, University of Illinois at Urbana-Champaign, Illinois, United States of America
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Illinois, United States of America
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Illinois, United States of America
| | - Amin Emad
- Department of Electrical and Computer Engineering, McGill University, Canada
| |
Collapse
|
145
|
Nishioka S, Wu PH, Yakabe T, Giaccia AJ, Le QT, Aoyama H, Shimizu S, Shirato H, Onodera Y, Nam JM. Rab27b contributes to radioresistance and exerts a paracrine effect via epiregulin in glioblastoma. Neurooncol Adv 2020; 2:vdaa091. [PMID: 33409495 PMCID: PMC7770522 DOI: 10.1093/noajnl/vdaa091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Radiotherapy is the standard treatment for glioblastoma (GBM). However, radioresistance of GBM cells leads to recurrence and poor patient prognosis. Recent studies suggest that secretion factors have important roles in radioresistance of tumor cells. This study aims to determine whether Rab27b, a small GTPase involved in secretory vesicle trafficking, plays a role in radioresistance of GBM. METHODS Microarray analysis, cell viability analysis, apoptosis assay, immunostaining, and in vivo experiments were performed to assess the effect of Rab27b on radioresistance of GBM. We further investigated paracrine effects mediated by Rab27b after X-ray irradiation using coculture systems of glioma cell lines. RESULTS Rab27b was specifically upregulated in irradiated U87MG cells. Furthermore, Rab27b knockdown decreased the proliferation of GBM cells after irradiation. Knockdown of Rab27b in U87MG cells combined with radiation treatment suppressed orthotopic tumor growth in the mouse brain and prolonged the survival of recipient mice. Interestingly, the co-upregulation of Rab27b and epiregulin (EREG), a member of the epidermal growth factor (EGF) family, correlated with radioresistance in glioma cell lines. Additionally, EREG, which was secreted from U87MG cells via Rab27b-mediated mechanism, activated EGF receptor and contributed to H4 cell proliferation in a paracrine manner. CONCLUSIONS Our results show that Rab27b mediates the radioresistance of highly malignant GBM cells. Rab27b promotes the proliferation of adjacent cells through EREG-mediated paracrine signaling after irradiation. Thus, the Rab27b-EREG pathway is a novel potential target to improve the efficacy of radiotherapy in GBM.
Collapse
Affiliation(s)
- Soichiro Nishioka
- Molecular and Cellular Dynamics Research, Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo, Japan
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Ping-Hsiu Wu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | | | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Quynh-Thu Le
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, California, USA
| | - Hidefumi Aoyama
- Department of Radiation Oncology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinichi Shimizu
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Radiation Medical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroki Shirato
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuhito Onodera
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
- Department of Molecular Biology, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Jin-Min Nam
- Global Center for Biomedical Science and Engineering, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
146
|
Abstract
The pathological features of the appendix tumors fundamentally recall those of the more frequent colorectal neoplasms, although with a higher relative incidence of carcinoids, due to the abundant presence of enteroendocrine cells in the appendix wall. Moreover, different types of lymphomas, Hodgkin and non-Hodgkin, arising from the extra-nodal mucosal-associated lymphatic tissue, can be encountered. The appendix tumor microenvironment (TME) consists of a cellular component and of a noncellular component: the former includes the immunocompetent cells, while the latter represents the support stroma. Particularly in carcinoids, the immune cell reaction can be explicated by tumor-infiltrating lymphocytes, which, in some circumstances, may arrange around and inside the tumor in a brisk fashion influencing favorably the prognosis. This active reaction has to be distinguished from any preexisting inflammatory condition of the appendix and from superimposed tumor complications, such as infection or ischemia. In practice, we consider the appendix TME a complex framework with immunological, mechanic, and metabolic functions, all supported by a marked neo-lymphoangiogenesis.
Collapse
|
147
|
Arrighetti N, Corbo C, Evangelopoulos M, Pastò A, Zuco V, Tasciotti E. Exosome-like Nanovectors for Drug Delivery in Cancer. Curr Med Chem 2019; 26:6132-6148. [PMID: 30182846 DOI: 10.2174/0929867325666180831150259] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/20/2018] [Accepted: 07/04/2018] [Indexed: 02/07/2023]
Abstract
Cancer treatment still represents a formidable challenge, despite substantial advancements in available therapies being made over the past decade. One major issue is poor therapeutic efficacy due to lack of specificity and low bioavailability. The progress of nanotechnology and the development of a variety of nanoplatforms have had a significant impact in improving the therapeutic outcome of chemotherapeutics. Nanoparticles can overcome various biological barriers and localize at tumor site, while simultaneously protecting a therapeutic cargo and increasing its circulation time. Despite this, due to their synthetic origin, nanoparticles are often detected by the immune system and preferentially sequestered by filtering organs. Exosomes have recently been investigated as suitable substitutes for the shortcomings of nanoparticles due to their biological compatibility and particularly small size (i.e., 30-150 nm). In addition, exosomes have been found to play important roles in cell communication, acting as natural carriers of biological cargoes throughout the body. This review aims to highlight the use of exosomes as drug delivery vehicles for cancer and showcases the various attempts used to exploit exosomes with a focus on the delivery of chemotherapeutics and nucleic acids.
Collapse
Affiliation(s)
- Noemi Arrighetti
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Claudia Corbo
- Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA, United States
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States
| | - Anna Pastò
- Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Valentina Zuco
- Department of Applied Research and Technological Development, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, 20133 Milan, Italy
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, 6670 Bertner Ave., Houston, TX, United States.,Houston Methodist Orthopedics & Sports Medicine, Houston Methodist Hospital, 6565 Fannin Street, Houston, TX, United States
| |
Collapse
|
148
|
Andreucci E, Ruzzolini J, Peppicelli S, Bianchini F, Laurenzana A, Carta F, Supuran CT, Calorini L. The carbonic anhydrase IX inhibitor SLC-0111 sensitises cancer cells to conventional chemotherapy. J Enzyme Inhib Med Chem 2019; 34:117-123. [PMID: 30362384 PMCID: PMC6211231 DOI: 10.1080/14756366.2018.1532419] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 02/07/2023] Open
Abstract
Drug combination represents one of the most accredited strategies of cancer therapy able to improve drug efficacy and possibly overcome drug resistance. Among the agents used to complement conventional chemotherapy, carbonic anhydrase IX (CAIX) inhibitors appear as one of the most suitable, as markers of hypoxic and acidic cancer cells which do not respond to chemo- and radiotherapy. We performed preclinical in vitro assays to evaluate whether the SLC-0111 CAIX inhibitor co-operates and potentiates the cytotoxic effects of conventional chemotherapeutic drugs in A375-M6 melanoma cells, MCF7 breast cancer cells, and HCT116 colorectal cancer cells. Here, we demonstrate that the SLC-0111 CAIX inhibitor potentiates cytotoxicity of Dacarbazine and Temozolomide currently used for advanced melanoma treatment. SLC-0111 also increases breast cancer cell response to Doxorubicin and enhances 5-Fluorouracil cytostatic activity on colon cancer cells. These findings disclose the possibility to extend the use of CAIX inhibitors in the combination therapy of various cancer histotypes.
Collapse
MESH Headings
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Antineoplastic Combined Chemotherapy Protocols/chemistry
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Carbonic Anhydrase IX/antagonists & inhibitors
- Carbonic Anhydrase IX/genetics
- Carbonic Anhydrase IX/metabolism
- Carbonic Anhydrase Inhibitors/chemistry
- Carbonic Anhydrase Inhibitors/pharmacology
- Cell Death/drug effects
- Cell Proliferation/drug effects
- Dacarbazine/analogs & derivatives
- Dacarbazine/chemistry
- Dacarbazine/pharmacology
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Fluorouracil/chemistry
- Fluorouracil/pharmacology
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- HCT116 Cells
- Humans
- MCF-7 Cells
- Molecular Structure
- Phenylurea Compounds/chemistry
- Phenylurea Compounds/pharmacology
- RNA, Messenger/antagonists & inhibitors
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Structure-Activity Relationship
- Sulfonamides/chemistry
- Sulfonamides/pharmacology
- Temozolomide
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Elena Andreucci
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Jessica Ruzzolini
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Silvia Peppicelli
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Francesca Bianchini
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Anna Laurenzana
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - Fabrizio Carta
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | | | - Lido Calorini
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
- Center of Excellence for Research, Transfer and High Education DenoTHE, University of Florence, Florence, Italy
| |
Collapse
|
149
|
Ngamcherdtrakul W, Yantasee W. siRNA therapeutics for breast cancer: recent efforts in targeting metastasis, drug resistance, and immune evasion. Transl Res 2019; 214:105-120. [PMID: 31487500 PMCID: PMC6848785 DOI: 10.1016/j.trsl.2019.08.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/22/2019] [Accepted: 08/15/2019] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA) has an established and precise mode of action to achieve protein knockdown. With the ability to target any protein, it is very attractive as a potential therapeutic for a plethora of diseases driven by the (over)expression of certain proteins. Utilizing siRNA to understand and treat cancer, a disease largely driven by genetic aberration, is thus actively investigated. However, the main hurdle for the clinical translation of siRNA therapeutics is to achieve effective delivery of siRNA molecules to tumors and the site of action, the cytosol, within cancer cells. Several nanoparticle delivery platforms for siRNA have been developed. In this Review, we describe recent efforts in developing siRNA therapeutics for the treatment of cancer, with particular emphasis on breast cancer. Instead of conventionally targeting proliferation and apoptosis aspects of tumorigenesis, we focus on recent attempts in targeting cancer's metastasis, drug resistance, and immune evasion, which are considered more challenging and less manageable in clinics with current therapeutic molecules. siRNA can target all proteins, including traditionally undruggable proteins, and is thus poised to address these clinical challenges. Evidence also suggests that siRNA can be superior to antibodies or small molecule inhibitors when inhibiting the same druggable pathway. In addition to cancer cells, the role of the tumor microenvironment has been increasingly appreciated. Components in the tumor microenvironment, particularly immune cells, and thus siRNA-based immunotherapy, are under extensive investigation. Lastly, multiple siRNAs with or without additional drugs can be codelivered on the same nanoparticle to the same target site of action, maximizing their potential synergy while limiting off-target toxicity.
Collapse
Affiliation(s)
| | - Wassana Yantasee
- PDX Pharmaceuticals, LLC, Portland, Oregon; Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon.
| |
Collapse
|
150
|
Emerging role of Immune Checkpoint Inhibitors in Hepatocellular Carcinoma. ACTA ACUST UNITED AC 2019; 55:medicina55100698. [PMID: 31627433 PMCID: PMC6843273 DOI: 10.3390/medicina55100698] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/07/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma is the most common primary liver cancer and the fourth leading cause of cancer death worldwide. A total of 70–80% of patients are diagnosed at an advanced stage with a dismal prognosis. Sorafenib had been the standard care for almost a decade until 2018 when the Food and Drug Administration approved an alternative first-line agent namely lenvatinib. Cabozantinib, regorafenib, and ramucirumab also displayed promising results in second line settings. FOLFOX4, however, results in an alternative first-line treatment for the Chinese clinical oncology guidelines. Moreover, nivolumab and pembrolizumab, two therapeutics against the Programmed death (PD)-ligand 1 (PD-L1)/PD1 axis have been recently approved for subsequent-line therapy. However, similar to other solid tumors, the response rate of single agent targeting PD-L1/PD1 axis is low. Therefore, a lot of combinatory approaches are under investigation, including the combination of different immune checkpoint inhibitors (ICIs), the addition of ICIs after resection or during loco-regional therapy, ICIs in addition to kinase inhibitors, anti-angiogenic therapeutics, and others. This review focuses on the use of ICIs for the hepatocellular carcinoma with a careful assessment of new ICIs-based combinatory approaches.
Collapse
|