1
|
Toulany M. Targeting K-Ras-mediated DNA damage response in radiation oncology: Current status, challenges and future perspectives. Clin Transl Radiat Oncol 2022; 38:6-14. [PMID: 36313934 PMCID: PMC9596599 DOI: 10.1016/j.ctro.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/06/2022] Open
Abstract
Approximately 60% of cancer patients receive curative or palliative radiation. Despite the significant role of radiotherapy (RT) as a curative approach for many solid tumors, tumor recurrence occurs, partially because of intrinsic radioresistance. Accumulating evidence indicates that the success of RT is hampered by activation of the DNA damage response (DDR). The intensity of DDR signaling is affected by multiple parameters, e.g., loss-of-function mutations in tumor suppressor genes, gain-of-function mutations in protooncogenes as well as radiation-induced alterations in signal-transduction pathways. Therefore, the response to irradiation differs in tumors of different types, which makes the individualization of RT as a rational but challenging goal. One contributor to tumor cell radiation survival is signaling through the Ras pathway. Three RAS genes encode 4 Ras isoforms: K-Ras4A, K-Ras4B, H-Ras, and N-Ras. RAS family members are found to be mutated in approximately 19% of human cancers. Mutations in RAS lead to constitutive activation of the gene product and activation of multiple Ras-dependent signal-transduction cascades. Preclinical studies have shown that the expression of mutant KRAS affects DDR and increases cell survival after irradiation. Approximately 70% of RAS mutations occur in KRAS. Thus, applying targeted therapies directly against K-Ras as well as K-Ras upstream activators and downstream effectors might be a tumor-specific approach to overcome K-Ras-mediated RT resistance. In this review, the role of K-Ras in the activation of DDR signaling will be summarized. Recent progress in targeting DDR in KRAS-mutated tumors in combination with radiochemotherapy will be discussed.
Collapse
|
2
|
Cayetano-Salazar L, Nava-Tapia DA, Astudillo-Justo KD, Arizmendi-Izazaga A, Sotelo-Leyva C, Herrera-Martinez M, Villegas-Comonfort S, Navarro-Tito N. Flavonoids as regulators of TIMPs expression in cancer: Consequences, opportunities, and challenges. Life Sci 2022; 308:120932. [PMID: 36067841 DOI: 10.1016/j.lfs.2022.120932] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 12/14/2022]
Abstract
Cancer is one of the leading causes of death in patients worldwide, where invasion and metastasis are directly responsible for this statement. Although cancer therapy has progressed in recent years, current therapeutic approaches are ineffective due to toxicity and chemoresistance. Therefore, it is essential to evaluate other treatment options, and natural products are a promising alternative as they show antitumor properties in different study models. This review describes the regulation of tissue inhibitors of metalloproteinases (TIMPs) expression and the role of flavonoids as molecules with the antitumor activity that targets TIMPs therapeutically. These inhibitors regulate tissue extracellular matrix (ECM) turnover; they inhibit matrix metalloproteinases (MMPs), cell migration, invasion, and angiogenesis and induce apoptosis in tumor cells. Data obtained in cell lines and in vivo models suggest that flavonoids are chemopreventive and cytotoxic against various types of cancer through several mechanisms. Flavonoids also regulate crucial signaling pathways such as focal adhesion kinase (FAK), phosphatidylinositol-3-kinase (PI3K)-Akt, signal transducer and activator of transcription 3 (STAT3), nuclear factor κB (NFκB), and mitogen-activated protein kinase (MAPK) involved in cancer cell migration, invasion, and metastasis. All these data reposition flavonoids as excellent candidates for use in cancer therapy.
Collapse
Affiliation(s)
- Lorena Cayetano-Salazar
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Dania A Nava-Tapia
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Kevin D Astudillo-Justo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Adán Arizmendi-Izazaga
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - César Sotelo-Leyva
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico
| | - Mayra Herrera-Martinez
- Instituto de Farmacobiología, Universidad de la Cañada, Teotitlán de Flores Magón, OAX 68540, Mexico
| | - Sócrates Villegas-Comonfort
- División de Ciencias Naturales e Ingeniería, Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, CDMX 05348, Mexico
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| |
Collapse
|
3
|
Fleming Martinez AK, Döppler HR, Bastea LI, Edenfield BH, Liou GY, Storz P. Ym1 + macrophages orchestrate fibrosis, lesion growth, and progression during development of murine pancreatic cancer. iScience 2022; 25:104327. [PMID: 35602933 PMCID: PMC9118688 DOI: 10.1016/j.isci.2022.104327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/04/2022] [Accepted: 04/26/2022] [Indexed: 01/05/2023] Open
Abstract
Desmoplasia around pancreatic lesions is a barrier for immune cells and a hallmark of developing and established pancreatic cancer. However, the contribution of the innate immune system to this process is ill-defined. Using the KC mouse model and primary cells in vitro, we show that alternatively activated macrophages (AAM) crosstalk with pancreatic lesion cells and pancreatic stellate cells (PSCs) to mediate fibrosis and progression of lesions. TGFβ1 secreted by AAM not only drives activation of quiescent PSCs but also in activated PSCs upregulates expression of TIMP1, a factor previously shown as crucial in fibrosis. Once activated, PSCs auto-stimulate proliferation via CXCL12. Furthermore, we found that TIMP1/CD63 signaling mediates PanIN lesion growth and TGFβ1 contributes to a cadherin switch and drives structural collapse of lesions, indicating a potential progression step. Taken together, our data indicate TGFβ1 produced by Ym1+ AAM as a major driver of processes that initiate the development of pancreatic cancer.
Collapse
Affiliation(s)
| | - Heike R. Döppler
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Ligia I. Bastea
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Brandy H. Edenfield
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| | - Geou-Yarh Liou
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA,Department of Biological Sciences, Center for Cancer Research & Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA,Corresponding author
| |
Collapse
|
4
|
Butera G, Brandi J, Cavallini C, Scarpa A, Lawlor RT, Scupoli MT, Marengo E, Cecconi D, Manfredi M, Donadelli M. The Mutant p53-Driven Secretome Has Oncogenic Functions in Pancreatic Ductal Adenocarcinoma Cells. Biomolecules 2020; 10:biom10060884. [PMID: 32526853 PMCID: PMC7356389 DOI: 10.3390/biom10060884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 05/30/2020] [Accepted: 06/05/2020] [Indexed: 02/06/2023] Open
Abstract
The cancer secretome is a rich repository of useful information for both cancer biology and clinical oncology. A better understanding of cancer secretome is particularly relevant for pancreatic ductal adenocarcinoma (PDAC), whose extremely high mortality rate is mainly due to early metastasis, resistance to conventional treatments, lack of recognizable symptoms, and assays for early detection. TP53 gene is a master transcriptional regulator controlling several key cellular pathways and it is mutated in ~75% of PDACs. We report the functional effect of the hot-spot p53 mutant isoforms R175H and R273H on cancer cell secretome, showing their influence on proliferation, chemoresistance, apoptosis, and autophagy, as well as cell migration and epithelial-mesenchymal transition. We compared the secretome of p53-null AsPC-1 PDAC cells after ectopic over-expression of R175H-mutp53 or R273H-mutp53 to identify the differentially secreted proteins by mutant p53. By using high-resolution SWATH-MS technology, we found a great number of differentially secreted proteins by the two p53 mutants, 15 of which are common to both mutants. Most of these secreted proteins are reported to promote cancer progression and epithelial-mesenchymal transition and might constitute a biomarker secreted signature that is driven by the hot-spot p53 mutants in PDAC.
Collapse
Affiliation(s)
- Giovanna Butera
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
| | - Jessica Brandi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Chiara Cavallini
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Aldo Scarpa
- Department of Diagnostics and Public health, Section of Pathology, University of Verona, 37134 Verona, Italy;
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Rita T. Lawlor
- ARC-Net Centre for Applied Research on Cancer, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
- Research Center LURM (Interdepartmental Laboratory of Medical Research), University of Verona, 37134 Verona, Italy;
| | - Emílio Marengo
- Department of Sciences and Technological Innovation, University of Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Italy, ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
| | - Daniela Cecconi
- Department of Biotechnology, University of Verona, 37134 Verona, Italy; (J.B.); (D.C.)
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Diseases, University of Piemonte Orientale, Italy, ISALIT, Spin-off at the University of Piemonte Orientale, 28100 Novara, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Italy, CAAD, corso Trieste 15/A, 28100 Novara, Italy
- Correspondence: (M.M.); (M.D.); Tel.: +39-032-1660810 (M.M.); +39-045-8027281 (M.D.); Fax: +39-045-8027170 (M.D.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences, Section of Biochemistry, University of Verona, Strada Le Grazie 8, 37134 Verona, Italy; (G.B.); (M.T.S.)
- Correspondence: (M.M.); (M.D.); Tel.: +39-032-1660810 (M.M.); +39-045-8027281 (M.D.); Fax: +39-045-8027170 (M.D.)
| |
Collapse
|
5
|
Tan Y, Li X, Tian Z, Chen S, Zou J, Lian G, Chen S, Huang K, Chen Y. TIMP1 down-regulation enhances gemcitabine sensitivity and reverses chemoresistance in pancreatic cancer. Biochem Pharmacol 2020; 189:114085. [PMID: 32522594 DOI: 10.1016/j.bcp.2020.114085] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/22/2022]
Abstract
The therapeutic effect of gemcitabine (GEM) in pancreatic ductal adenocarcinoma (PDAC) is limited due to low drug sensitivity and high drug resistance. Tissue inhibitor of matrix metalloprotease 1 (TIMP1) is reportedly associated with GEM resistance in PDAC. However, the effect of TIMP1 down-regulation in combination with GEM treatment is unknown. We analyzed the expression of TIMP1 in human PDAC tissue using western blot, quantitative real-time polymerase chain reaction (qRT-PCR), and immunohistochemistry. TIMP1 was highly expressed in PDAC specimens. Kaplan-Meier survival analysis suggested that a higher level of TIMP1 was correlated with poorer overall survival in 103 PDAC patients. The mRNA and protein expression profiles of TIMP1 were explored in the HTERT-HPNE human pancreatic ductal epithelium cell line, five PDAC cell lines (MIA PaCa-2, PANC-1, BxPC-3, Capan2, and SW1990), and two GEM-resistant PDAC cell lines (MIA PaCa-2R and PANC-1R). Compared with HTERT-HPNE, TIMP1 was highly expressed in the PDAC cell lines. In addition, TIMP1 was upregulated in GEM-resistant PDAC cell lines compared with their parental cells. When TIMP1 was knocked-down using short hairpin RNA, GEM-induced cytotoxicity and apoptosis were increased, while colony formation was repressed in MIA PaCa-2, PANC-1, and their GEM-resistant cells. When Bax was activated by BAM7 or Bcl-2 was inhibited by venetoclax, CCK-8 assays demonstrated that GEM sensitivity was restored in GEM-resistant cells. When Bax was down-regulated by siRNA, CCK-8 assays verified that GEM sensitivity was decreased in PDAC cells. The observations that TIMP1 knockdown enhanced GEM sensitivity and reversed chemoresistance by inducing cells apoptosis indicated cooperative antitumor effects of shTIMP1 and GEM therapy on PDAC cells. The combination may be a potential strategy for PDAC therapy.
Collapse
Affiliation(s)
- Ying Tan
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xuanna Li
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhenfeng Tian
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shangxiang Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jinmao Zou
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Guoda Lian
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shaojie Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kaihong Huang
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| | - Yinting Chen
- Department of Gastroenterology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Nordgaard C, Doll S, Matos ALDSA, Høeberg M, Kazi JU, Friis S, Stenvang J, Rönnstrand L, Mann M, Manuel Afonso Moreira J. Metallopeptidase inhibitor 1 (TIMP-1) promotes receptor tyrosine kinase c-Kit signaling in colorectal cancer. Mol Oncol 2019; 13:2646-2662. [PMID: 31545548 PMCID: PMC6887592 DOI: 10.1002/1878-0261.12575] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) is the third most prevalent cancer worldwide causing an estimated 700 000 deaths annually. Different types of treatment are available for patients with advanced metastatic colorectal cancer, including targeted biological agents, such as cetuximab, a monoclonal antibody that targets EGFR. We have previously reported a study indicating multiple levels of interaction between metallopeptidase inhibitor 1 (TIMP‐1) and the epidermal growth factor (EGF) signaling axis, which could explain how TIMP‐1 levels can affect the antitumor effects of EGFR inhibitors. We also reported an association between TIMP‐1‐mediated cell invasive behavior and KRAS status. To gain insight into the molecular mechanisms underlying the effects of TIMP‐1 in CRC, we examined by transcriptomics, proteomics, and kinase activity profiling a matched pair of isogenic human CRC isogenic DLD‐1 CRC cell clones, bearing either an hemizygous KRAS wild‐type allele or KRAS G13D mutant allele, exposed, or not, to TIMP‐1. Omics analysis of the two cell lines identified the receptor tyrosine kinase c‐Kit, a proto‐oncogene that can modulate cell proliferation and invasion in CRC, as a target for TIMP‐1. We found that exposure of DLD‐1 CRC cells to exogenously added TIMP‐1 promoted phosphorylation of c‐Kit, indicative of a stimulatory effect of TIMP‐1 on the c‐Kit signaling axis. In addition, TIMP‐1 inhibited c‐Kit shedding in CRC cells grown in the presence of exogenous TIMP‐1. Given the regulatory roles that c‐Kit plays in cell proliferation and migration, and the realization that c‐Kit is an important oncogene in CRC, it is likely that some of the biological effects of TIMP‐1 overexpression in CRC may be exerted through its effect on c‐Kit signaling.
Collapse
Affiliation(s)
- Cathrine Nordgaard
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Sophia Doll
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | - Mikkel Høeberg
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Julhash Uddin Kazi
- Division of Translational Cancer Research and Lund Stem Cell Center, Lund University, Sweden
| | - Stine Friis
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Jan Stenvang
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Lars Rönnstrand
- Division of Translational Cancer Research and Lund Stem Cell Center, Lund University, Sweden.,Division of Oncology, Skåne University Hospital, Lund, Sweden
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.,Novo Nordisk Foundation Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - José Manuel Afonso Moreira
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
7
|
Malik R, Luong T, Cao X, Han B, Shah N, Franco-Barraza J, Han L, Shenoy VB, Lelkes PI, Cukierman E. Rigidity controls human desmoplastic matrix anisotropy to enable pancreatic cancer cell spread via extracellular signal-regulated kinase 2. Matrix Biol 2019; 81:50-69. [PMID: 30412725 PMCID: PMC6504628 DOI: 10.1016/j.matbio.2018.11.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/26/2018] [Accepted: 11/01/2018] [Indexed: 02/06/2023]
Abstract
It is predicted that pancreatic ductal adenocarcinoma (PDAC) will become the second most lethal cancer in the US by 2030. PDAC includes a fibrous-like stroma, desmoplasia, encompassing most of the tumor mass, which is produced by cancer-associated fibroblasts (CAFs) and includes their cell-derived extracellular matrices (CDMs). Since elimination of desmoplasia has proven detrimental to patients, CDM reprogramming, as opposed to stromal ablation, is therapeutically desirable. Hence, efforts are being made to harness desmoplasia's anti-tumor functions. We conducted biomechanical manipulations, using variations of pathological and physiological substrates in vitro, to culture patient-harvested CAFs and generate CDMs that restrict PDAC growth and spread. We posited that extrinsic modulation of the environment, via substrate rigidity, influences CAF's cell-intrinsic forces affecting CDM production. Substrates used were polyacrylamide gels of physiological (~1.5 kPa) or pathological (~7 kPa) stiffnesses. Results showed that physiological substrates influenced CAFs to generate CDMs similar to normal/control fibroblasts. We found CDMs to be softer than the corresponding underlying substrates, and CDM fiber anisotropy (i.e., alignment) to be biphasic and informed via substrate-imparted morphological CAF aspect ratios. The biphasic nature of CDM fiber anisotropy was mathematically modeled and proposed a correlation between CAF aspect ratios and CDM alignment; regulated by extrinsic and intrinsic forces to conserve minimal free energy. Biomechanical manipulation of CDMs, generated on physiologically soft substrates, leads to reduction in nuclear translocation of pERK1/2 in KRAS mutated pancreatic cells. ERK2 was found essential for CDM-regulated tumor cell spread. In vitro findings correlated with in vivo observations; nuclear pERK1/2 is significantly high in human PDAC samples. The study suggests that altering underlying substrates enable CAFs to remodel CDMs and restrict pancreatic cancer cell spread in an ERK2 dependent manner.
Collapse
Affiliation(s)
- R Malik
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, United States of America; Department Bioengineering, Temple University, United States of America
| | - T Luong
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, United States of America
| | - X Cao
- Materials Science and Engineering, University of Pennsylvania, United States of America
| | - B Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States of America
| | - N Shah
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, United States of America
| | - J Franco-Barraza
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, United States of America
| | - L Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, United States of America
| | - V B Shenoy
- Materials Science and Engineering, University of Pennsylvania, United States of America
| | - P I Lelkes
- Department Bioengineering, Temple University, United States of America.
| | - E Cukierman
- Cancer Biology Program, Marvin & Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, United States of America.
| |
Collapse
|
8
|
Khader S, Thyagarajan A, Sahu RP. Exploring Signaling Pathways and Pancreatic Cancer Treatment Approaches Using Genetic Models. Mini Rev Med Chem 2019; 19:1112-1125. [PMID: 30924420 DOI: 10.2174/1389557519666190327163644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/12/2019] [Accepted: 03/19/2019] [Indexed: 11/22/2022]
Abstract
Despite available treatment options, the overall survival rates of pancreatic cancer patients remain dismal. Multiple counter-regulatory pathways have been identified and shown to be involved in interfering with the efficacy of therapeutic agents. In addition, various known genetic alterations in the cellular signaling pathways have been implicated in affecting the growth and progression of pancreatic cancer. Nevertheless, the significance of other unknown pathways is yet to be explored, which provides the rationale for the intervention of new approaches. Several experimental genetic models have been explored to define the impact of key signaling cascades, and their mechanisms in the pathophysiology as well as treatment approaches of pancreatic cancer. The current review highlights the recent updates, and significance of such genetic models in the therapeutic efficacy of anti-tumor agents including the standard chemotherapeutic agents, natural products, cell signaling inhibitors, immunebased therapies and the combination of these approaches in pancreatic cancer.
Collapse
Affiliation(s)
- Shorooq Khader
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| | - Anita Thyagarajan
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| | - Ravi P Sahu
- Department of Pharmacology and Toxicology, Boonshoft School of Medicine at Wright State University, Dayton, OH 45345, United States
| |
Collapse
|
9
|
Tarpgaard LS, Ørum-Madsen MS, Christensen IJ, Nordgaard C, Noer J, Guren TK, Glimelius B, Sorbye H, Ikdahl T, Kure EH, Tveit KM, Nielsen HJ, Pfeiffer P, Brünner N, Moreira JMA. TIMP-1 is under regulation of the EGF signaling axis and promotes an aggressive phenotype in KRAS-mutated colorectal cancer cells: a potential novel approach to the treatment of metastatic colorectal cancer. Oncotarget 2018; 7:59441-59457. [PMID: 27509063 PMCID: PMC5312323 DOI: 10.18632/oncotarget.11118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/17/2016] [Indexed: 01/05/2023] Open
Abstract
It is now widely accepted that therapeutic antibodies targeting epidermal growth factor receptor (EGFR) can have efficacy in KRAS wild-type advanced colorectal cancer (CRC) patients. What remains to be ascertained is whether a subgroup of KRAS-mutated CRC patients might not also derive benefit from EGFR inhibitors. Metalloproteinase inhibitor 1 (TIMP-1) is a pleiotropic factor predictive of survival outcome of CRC patients. Levels of TIMP-1 were measured in pre-treatment plasma samples (n = 426) of metastatic CRC patients randomized to Nordic FLOX (5-fluorouracil and oxaliplatin) +/− cetuximab (NORDIC VII study). Multivariate analysis demonstrated a significant interaction between plasma TIMP-1 protein levels, KRAS status and treatment with patients bearing KRAS mutated tumors and high TIMP-1 plasma level (> 3rd quartile) showing a significantly longer overall survival if treated with cetuximab (HR, 0.48; 95% CI, 0.25 to 0.93). To gain mechanistic insights into this association we analyzed a set of five different CRC cell lines. We show here that EGFR signaling induces TIMP-1 expression in CRC cells, and that TIMP-1 promotes a more aggressive behavior, specifically in KRAS mutated cells. The two sets of data, clinical and in vitro, are complementary and support each other, lending strength to our contention that TIMP- 1 plasma levels can identify a subset of patients with KRAS-mutated metastatic CRC that will have benefit from EGFR-inhibition therapy.
Collapse
Affiliation(s)
- Line S Tarpgaard
- Department of Oncology, Odense University Hospital, Odense, Denmark and University of Southern Denmark, Odense, Denmark
| | - Maj Sofie Ørum-Madsen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ib J Christensen
- The Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark and Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Cathrine Nordgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Julie Noer
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tormod K Guren
- Department of Oncology and K. G. Jebsen Centre for Colorectal Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bengt Glimelius
- Departments of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala and Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Halfdan Sorbye
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.,Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Tone Ikdahl
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elin H Kure
- Department of Genetics, Oslo University Hospital, Oslo, Norway
| | - Kjell M Tveit
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Hans J Nielsen
- Department of Surgical Gastroenterology, Copenhagen University Hospital, Hvidovre, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark and University of Southern Denmark, Odense, Denmark
| | - Nils Brünner
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - José M A Moreira
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
HuoXueJieDu Formula Alleviates Diabetic Retinopathy in Rats by Inhibiting SOCS3-STAT3 and TIMP1-A2M Pathways. Int J Genomics 2017; 2017:4832125. [PMID: 29318137 PMCID: PMC5727685 DOI: 10.1155/2017/4832125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/30/2017] [Accepted: 10/08/2017] [Indexed: 02/07/2023] Open
Abstract
HuoXueJieDu (HXJD) formula exerts protective effects against diabetic retinopathy (DR) in rats, but its underlying mechanism remains unknown. In the present study, the diabetic rats were established using streptozocin. The administration of HXJD was initiated at 20 weeks after diabetes induction and continued for 12 weeks. Whole genome expression profiles in rat retinas were examined using microarray technology. Differential gene expression and pathway enrichment analysis were conducted on the microarray data, with validation through real-time PCR and immunohistochemical staining. The results showed that 170 genes and several IPA canonical pathways related to inflammation, matrix metabolism, and phototransduction were regulated by HXJD. PCR validation of selected genes, including SOCS3, STAT3, TIMP1, and A2M, confirmed the gene expression changes influenced by HXJD. In addition, the immunohistochemical staining results suggested that critical members of the SOCS3-STAT3 pathway were also affected by HXJD. Taken together, these results indicated that SOCS3-STAT3 and TIMP1-A2M pathways might mediate the alleviation of HXJD activities in rats with diabetic retinopathy.
Collapse
|
11
|
D'Costa Z, Jones K, Azad A, van Stiphout R, Lim SY, Gomes AL, Kinchesh P, Smart SC, Gillies McKenna W, Buffa FM, Sansom OJ, Muschel RJ, O'Neill E, Fokas E. Gemcitabine-Induced TIMP1 Attenuates Therapy Response and Promotes Tumor Growth and Liver Metastasis in Pancreatic Cancer. Cancer Res 2017; 77:5952-5962. [PMID: 28765154 DOI: 10.1158/0008-5472.can-16-2833] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 03/01/2017] [Accepted: 07/19/2017] [Indexed: 11/16/2022]
Abstract
Gemcitabine constitutes one of the backbones for chemotherapy treatment in pancreatic ductal adenocarcinoma (PDAC), but patients often respond poorly to this agent. Molecular markers downstream of gemcitabine treatment in preclinical models may provide an insight into resistance mechanisms. Using cytokine arrays, we identified potential secretory biomarkers of gemcitabine resistance (response) in the transgenic KRasG12D; Trp53R172H; Pdx-1 Cre (KPC) mouse model of PDAC. We verified the oncogenic role of the cytokine tissue inhibitor of matrix metalloproteinases 1 (TIMP1) in primary pancreatic tumors and metastases using both in vitro techniques and animal models. We identified potential pathways affected downstream of TIMP1 using the Illumina Human H12 array. Our findings were validated in both primary and metastatic models of pancreatic cancer. Gemcitabine increased inflammatory cytokines including TIMP1 in the KPC mouse model. TIMP1 was upregulated in patients with pancreatic intraepithelial neoplasias grade 3 and PDAC lesions relative to matched normal pancreatic tissue. In addition, TIMP1 played a role in tumor clonogenic survival and vascular density, while TIMP1 inhibition resensitized tumors to gemcitabine and radiotherapy. We observed a linear relationship between TIMP-1 expression, liver metastatic burden, and infiltration by CD11b+Gr1+ myeloid cells and CD4+CD25+FOXP3+ Tregs, whereas the presence of tumor cells was required for immune cell infiltration. Overall, our results identify TIMP1 upregulation as a resistance mechanism to gemcitabine and provide a rationale for combining chemo/radiotherapy with TIMP1 inhibitors in PDAC. Cancer Res; 77(21); 5952-62. ©2017 AACR.
Collapse
Affiliation(s)
- Zenobia D'Costa
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Keaton Jones
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Abul Azad
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Ruud van Stiphout
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Su Y Lim
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | | | - Paul Kinchesh
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Sean C Smart
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - W Gillies McKenna
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Francesca M Buffa
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Owen J Sansom
- CRUK Beatson Institute of Oncology, University of Glasgow, Glasgow, United Kingdom
| | - Ruth J Muschel
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom
| | - Eric O'Neill
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.
| | - Emmanouil Fokas
- Department of Oncology, CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
12
|
Capello M, Bantis LE, Scelo G, Zhao Y, Li P, Dhillon DS, Patel NJ, Kundnani DL, Wang H, Abbruzzese JL, Maitra A, Tempero MA, Brand R, Firpo MA, Mulvihill SJ, Katz MH, Brennan P, Feng Z, Taguchi A, Hanash SM. Sequential Validation of Blood-Based Protein Biomarker Candidates for Early-Stage Pancreatic Cancer. J Natl Cancer Inst 2017; 109:2952681. [PMID: 28376157 PMCID: PMC5441297 DOI: 10.1093/jnci/djw266] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/17/2016] [Accepted: 10/06/2016] [Indexed: 02/06/2023] Open
Abstract
Background CA19-9, which is currently in clinical use as a pancreatic ductal adenocarcinoma (PDAC) biomarker, has limited performance in detecting early-stage disease. We and others have identified protein biomarker candidates that have the potential to complement CA19-9. We have carried out sequential validations starting with 17 protein biomarker candidates to determine which markers and marker combination would improve detection of early-stage disease compared with CA19-9 alone. Methods Candidate biomarkers were subjected to enzyme-linked immunosorbent assay based sequential validation using independent multiple sample cohorts consisting of PDAC cases (n = 187), benign pancreatic disease (n = 93), and healthy controls (n = 169). A biomarker panel for early-stage PDAC was developed based on a logistic regression model. All statistical tests for the results presented below were one-sided. Results Six out of the 17 biomarker candidates and CA19-9 were validated in a sample set consisting of 75 PDAC patients, 27 healthy subjects, and 19 chronic pancreatitis patients. A second independent set of 73 early-stage PDAC patients, 60 healthy subjects, and 74 benign pancreatic disease patients (combined validation set) yielded a model that consisted of TIMP1, LRG1, and CA19-9. Additional blinded testing of the model was done using an independent set of plasma samples from 39 resectable PDAC patients and 82 matched healthy subjects (test set). The model yielded areas under the curve (AUCs) of 0.949 (95% confidence interval [CI] = 0.917 to 0.981) and 0.887 (95% CI = 0.817 to 0.957) with sensitivities of 0.849 and 0.667 at 95% specificity in discriminating early-stage PDAC vs healthy subjects in the combined validation and test sets, respectively. The performance of the biomarker panel was statistically significantly improved compared with CA19-9 alone (P < .001, combined validation set; P = .008, test set). Conclusion The addition of TIMP1 and LRG1 immunoassays to CA19-9 statistically significantly improves the detection of early-stage PDAC.
Collapse
Affiliation(s)
- Michela Capello
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Leonidas E. Bantis
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Ghislaine Scelo
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Yang Zhao
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Peng Li
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Dilsher S. Dhillon
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Nikul J. Patel
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Deepali L. Kundnani
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Hong Wang
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - James L. Abbruzzese
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Anirban Maitra
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Margaret A. Tempero
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Randall Brand
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Matthew A. Firpo
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Sean J. Mulvihill
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Matthew H. Katz
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Paul Brennan
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Ziding Feng
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Ayumu Taguchi
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| | - Samir M. Hanash
- Affiliations of authors: Departments of Clinical Cancer Prevention (MC, DSD, NJP, DLK, HW, SMH), Biostatistics (LEB, YZ, ZF), Pathology (AM), Surgical Oncology (MHK), and Translational Molecular Pathology (AT), The University of Texas MD Anderson Cancer Center, Houston, TX; International Agency for Research on Cancer (IARC), Lyon, France (GS, PL, PB); Division of Medical Oncology, Duke University, Durham, NC (JLA); Pancreas Center, University of California San Francisco, Helen Diller Family Comprehensive Cancer Center, San Francisco, CA (MAT); Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh, Pittsburgh, PA (RB); Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT (MAF, SJM)
| |
Collapse
|
13
|
Ye X, Chan KC, Waters AM, Bess M, Harned A, Wei BR, Loncarek J, Luke BT, Orsburn BC, Hollinger BD, Stephens RM, Bagni R, Martinko A, Wells JA, Nissley DV, McCormick F, Whiteley G, Blonder J. Comparative proteomics of a model MCF10A-KRasG12V cell line reveals a distinct molecular signature of the KRasG12V cell surface. Oncotarget 2016; 7:86948-86971. [PMID: 27894102 PMCID: PMC5341332 DOI: 10.18632/oncotarget.13566] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 11/07/2016] [Indexed: 11/25/2022] Open
Abstract
Oncogenic Ras mutants play a major role in the etiology of most aggressive and deadly carcinomas in humans. In spite of continuous efforts, effective pharmacological treatments targeting oncogenic Ras isoforms have not been developed. Cell-surface proteins represent top therapeutic targets primarily due to their accessibility and susceptibility to different modes of cancer therapy. To expand the treatment options of cancers driven by oncogenic Ras, new targets need to be identified and characterized at the surface of cancer cells expressing oncogenic Ras mutants. Here, we describe a mass spectrometry-based method for molecular profiling of the cell surface using KRasG12V transfected MCF10A (MCF10A-KRasG12V) as a model cell line of constitutively activated KRas and native MCF10A cells transduced with an empty vector (EV) as control. An extensive molecular map of the KRas surface was achieved by applying, in parallel, targeted hydrazide-based cell-surface capturing technology and global shotgun membrane proteomics to identify the proteins on the KRasG12V surface. This method allowed for integrated proteomic analysis that identified more than 500 cell-surface proteins found unique or upregulated on the surface of MCF10A-KRasG12V cells. Multistep bioinformatic processing was employed to elucidate and prioritize targets for cross-validation. Scanning electron microscopy and phenotypic cancer cell assays revealed changes at the cell surface consistent with malignant epithelial-to-mesenchymal transformation secondary to KRasG12V activation. Taken together, this dataset significantly expands the map of the KRasG12V surface and uncovers potential targets involved primarily in cell motility, cellular protrusion formation, and metastasis.
Collapse
Affiliation(s)
- Xiaoying Ye
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - King C. Chan
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Andrew M. Waters
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Matthew Bess
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Adam Harned
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Bih-Rong Wei
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Jadranka Loncarek
- Laboratory of Protein Dynamics and Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Brian T. Luke
- Advanced Biomedical Computing Center, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | | | - Bradley D. Hollinger
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Robert M. Stephens
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Rachel Bagni
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Alex Martinko
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158-2517, USA
| | - James A. Wells
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158-2517, USA
| | - Dwight V. Nissley
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Frank McCormick
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158-9001, USA
| | - Gordon Whiteley
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Josip Blonder
- Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| |
Collapse
|
14
|
Drosos Y, Neale G, Ye J, Paul L, Kuliyev E, Maitra A, Means AL, Washington MK, Rehg J, Finkelstein DB, Sosa-Pineda B. Prox1-Heterozygosis Sensitizes the Pancreas to Oncogenic Kras-Induced Neoplastic Transformation. Neoplasia 2016; 18:172-84. [PMID: 26992918 PMCID: PMC4796801 DOI: 10.1016/j.neo.2016.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/29/2016] [Accepted: 02/09/2016] [Indexed: 12/15/2022] Open
Abstract
The current paradigm of pancreatic neoplastic transformation proposes an initial step whereby acinar cells convert into acinar-to-ductal metaplasias, followed by progression of these lesions into neoplasias under sustained oncogenic activity and inflammation. Understanding the molecular mechanisms driving these processes is crucial to the early diagnostic and prevention of pancreatic cancer. Emerging evidence indicates that transcription factors that control exocrine pancreatic development could have either, protective or facilitating roles in the formation of preneoplasias and neoplasias in the pancreas. We previously identified that the homeodomain transcription factor Prox1 is a novel regulator of mouse exocrine pancreas development. Here we investigated whether Prox1 function participates in early neoplastic transformation using in vivo, in vitro and in silico approaches. We found that Prox1 expression is transiently re-activated in acinar cells undergoing dedifferentiation and acinar-to-ductal metaplastic conversion. In contrast, Prox1 expression is largely absent in neoplasias and tumors in the pancreas of mice and humans. We also uncovered that Prox1-heterozygosis markedly increases the formation of acinar-to-ductal-metaplasias and early neoplasias, and enhances features associated with inflammation, in mouse pancreatic tissues expressing oncogenic Kras. Furthermore, we discovered that Prox1-heterozygosis increases tissue damage and delays recovery from inflammation in pancreata of mice injected with caerulein. These results are the first demonstration that Prox1 activity protects pancreatic cells from acute tissue damage and early neoplastic transformation. Additional data in our study indicate that this novel role of Prox1 involves suppression of pathways associated with inflammatory responses and cell invasiveness.
Collapse
Affiliation(s)
- Yiannis Drosos
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Geoffrey Neale
- Department of Hartwell Center for Bioinformatics and Biotechnology, St. Jude Children's Research Hospital, Memphis, TN
| | - Jianming Ye
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Leena Paul
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Emin Kuliyev
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN
| | - Anirban Maitra
- The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anna L Means
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Jerold Rehg
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN
| | - David B Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN
| | - Beatriz Sosa-Pineda
- Department of Genetics, St. Jude Children's Research Hospital, Memphis, TN; Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL.
| |
Collapse
|
15
|
Li Q, Zhang CS, Zhang Y. Molecular aspects of prostate cancer with neuroendocrine differentiation. Chin J Cancer Res 2016; 28:122-9. [PMID: 27041934 DOI: 10.3978/j.issn.1000-9604.2016.01.02] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuroendocrine differentiation (NED), which is not uncommon in prostate cancer, is increases in prostate cancer after androgen-deprivation therapy (ADT) and generally appears in castration-resistant prostate cancer (CRPC). Neuroendocrine cells, which are found in normal prostate tissue, are a small subset of cells and have unique function in regulating the growth of prostate cells. Prostate cancer with NED includes different types of tumor, including focal NED, pure neuroendocrine tumor or mixed neuroendocrine-adenocarcinoma. Although more and more studies are carried out on NED in prostate cancer, the molecular components that are involved in NED are still poorly elucidated. We review neuroendocrine cells in normal prostate tissue, NED in prostate cancer, terminology of NED and biomarkers used for detecting NED in routine pathological practice. Some recently reported molecular components which drive NED in prostate cancer are listed in the review.
Collapse
Affiliation(s)
- Qi Li
- 1 Department of Pathology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China ; 2 MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Connie S Zhang
- 1 Department of Pathology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China ; 2 MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yifen Zhang
- 1 Department of Pathology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China ; 2 MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
16
|
The tissue inhibitor of metalloproteinases 1 increases the clonogenic efficiency of human hematopoietic progenitor cells through CD63/PI3K/Akt signaling. Exp Hematol 2015. [DOI: 10.1016/j.exphem.2015.07.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
17
|
Cancer subclonal genetic architecture as a key to personalized medicine. Neoplasia 2014; 15:1410-20. [PMID: 24403863 DOI: 10.1593/neo.131972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/03/2013] [Accepted: 12/03/2013] [Indexed: 02/08/2023] Open
Abstract
The future of personalized oncological therapy will likely rely on evidence-based medicine to integrate all of the available evidence to delineate the most efficacious treatment option for the patient. To undertake evidence-based medicine through use of targeted therapy regimens, identification of the specific underlying causative mutation(s) driving growth and progression of a patient's tumor is imperative. Although molecular subtyping is important for planning and treatment, intraclonal genetic diversity has been recently highlighted as having significant implications for biopsy-based prognosis. Overall, delineation of the clonal architecture of a patient's cancer and how this will impact on the selection of the most efficacious therapy remain a topic of intense interest.
Collapse
|
18
|
Ki CS, Shih H, Lin CC. Effect of 3D matrix compositions on the efficacy of EGFR inhibition in pancreatic ductal adenocarcinoma cells. Biomacromolecules 2013; 14:3017-26. [PMID: 23889305 DOI: 10.1021/bm4004496] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Therapeutics to inhibit signaling of epidermal growth factor receptor (EGFR) has been suggested as a potential treatment for pancreatic cancers, and two-dimensional (2D) cell culture techniques are commonly used to identify and/or verify the therapeutic efficacy of EGFR inhibitors. However, drug targets identified from conventional cell culture techniques may not exhibit desired functions when these drugs are tested in animal studies, in large part due to the complicated tumor microenvironments. Hence, it is crucial to develop a biomimetic cell culture system capable of recapitulating aspects of tumor niches for studying cancer cell fate processes under the influence of various environmental stimuli. In this study, we utilized a versatile PEG-peptide hydrogel system to demonstrate the influence of matrix properties and EGFR inhibition on the growth of a pancreatic ductal adenocarcinoma cell line (PANC-1). PANC-1 cells were encapsulated in 8-arm PEG-norbornene (PEG8NB) hydrogels cross-linked by matrix metalloproteinase (MMP) sensitive peptide (MMP(Linker)) using thiol-ene photoclick chemistry. In soft hydrogels (G' ~ 2 kPa), cells retained high initial viability and formed clusters after prolonged culture, whereas cells encapsulated in stiff hydrogels (G' ~ 12 kPa) exhibited lower initial viability and reduced proliferation. While the immobilization of an EGFR peptide inhibitor, Asn-Tyr-Gln-Gln-Asn or NYQQN, in soft hydrogels did not cause cell death, this peptide induced significant cell apoptosis when immobilized in stiff hydrogels. Western blotting results showed that cell death was due to reduced expression of EGFR and Akt in stiff hydrogels under the influence of immobilized NYQQN peptide. These results shed light on the importance and non-negligible role of matrix properties on the efficacy of antitumor drugs.
Collapse
Affiliation(s)
- Chang Seok Ki
- Department of Biomedical Engineering, Purdue School of Engineering and Technology, Indiana University-Purdue University at Indianapolis, Indianapolis, Indiana 46202, United States
| | | | | |
Collapse
|