1
|
Awale S, Maneenet J, Phan ND, Nguyen HH, Fujii T, Ihmels H, Soost D, Tajuddeen N, Feineis D, Bringmann G. Toyaburgine, a Synthetic N-Biphenyl-Dihydroisoquinoline Inspired by Related N, C-Coupled Naphthylisoquinoline Alkaloids, with High In Vivo Efficacy in Preclinical Pancreatic Cancer Models. ACS Chem Biol 2025; 20:917-929. [PMID: 40048237 DOI: 10.1021/acschembio.4c00870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Pancreatic cancer is a highly aggressive and lethal malignancy, with a 5-year survival rate below 10%. Traditional chemotherapy, including gemcitabine, has limited efficacy due to chemoresistance and a unique tumor microenvironment characterized by hypovascularity and nutrient deprivation. This study reports on the discovery of a new N-biphenyl-dihydroisoquinoline, named toyaburgine (4), inspired by naturally occurring N,C-coupled naphthylisoquinoline alkaloids. Developed through systematic structural optimization, toyaburgine is a potent anticancer agent, showing promise for pancreatic cancer treatment. It exhibits strong antiausterity activity with low nanomolar PC50 values, effectively inhibiting pancreatic cancer cell viability under nutrient-deprived conditions. In vitro, 4 causes significant morphological changes and cancer cell death in MIA PaCa-2 cells while also inhibiting cell migration and colony formation, which indicates its antimetastatic potential. Mechanistically, toyaburgine disrupts the PI3K/Akt/mTOR pathway, essential for pancreatic cancer cell survival in a stressful microenvironment, and inhibits MIA PaCa-2 spheroid formation. In vivo, toyaburgine, alone or combined with gemcitabine, shows effective tumor suppression in subcutaneous xenograft and clinically relevant orthotopic models, where it also reduces cachexia. These results highlight the potential of toyaburgine as a new therapeutic drug for pancreatic cancer. Its combination with gemcitabine presents a promising treatment approach by targeting both proliferating and gemcitabine-resistant cancer cells.
Collapse
Affiliation(s)
- Suresh Awale
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Juthamart Maneenet
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Nguyen Duy Phan
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Hung Hong Nguyen
- Natural Drug Discovery Laboratory, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Tsutomu Fujii
- Department of Surgery and Science, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Heiko Ihmels
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Denisa Soost
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
| | - Nasir Tajuddeen
- Department of Chemistry-Biology, and Center of Micro- and Nanochemistry and (Bio)Technology (Cμ), University of Siegen, Adolf-Reichwein-Straße 2, 57068 Siegen, Germany
- Department of Chemistry, Ahmadu Bello University, 810107 Zaria, Nigeria
| | - Doris Feineis
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Gerhard Bringmann
- Institute of Organic Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| |
Collapse
|
2
|
Bharadwaj AG, Kempster E, Waisman DM. The ANXA2/S100A10 Complex—Regulation of the Oncogenic Plasminogen Receptor. Biomolecules 2021; 11:biom11121772. [PMID: 34944416 PMCID: PMC8698604 DOI: 10.3390/biom11121772] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
The generation of the serine protease plasmin is initiated by the binding of its zymogenic precursor, plasminogen, to cell surface receptors. The proteolytic activity of plasmin, generated at the cell surface, plays a crucial role in several physiological processes, including fibrinolysis, angiogenesis, wound healing, and the invasion of cells through both the basement membrane and extracellular matrix. The seminal observation by Albert Fischer that cancer cells, but not normal cells in culture, produce large amounts of plasmin formed the basis of current-day observations that plasmin generation can be hijacked by cancer cells to allow tumor development, progression, and metastasis. Thus, the cell surface plasminogen-binding receptor proteins are critical to generating plasmin proteolytic activity at the cell surface. This review focuses on one of the twelve well-described plasminogen receptors, S100A10, which, when in complex with its regulatory partner, annexin A2 (ANXA2), forms the ANXA2/S100A10 heterotetrameric complex referred to as AIIt. We present the theme that AIIt is the quintessential cellular plasminogen receptor since it regulates the formation and the destruction of plasmin. We also introduce the term oncogenic plasminogen receptor to define those plasminogen receptors directly activated during cancer progression. We then discuss the research establishing AIIt as an oncogenic plasminogen receptor-regulated during EMT and activated by oncogenes such as SRC, RAS, HIF1α, and PML-RAR and epigenetically by DNA methylation. We further discuss the evidence derived from animal models supporting the role of S100A10 in tumor progression and oncogenesis. Lastly, we describe the potential of S100A10 as a biomarker for cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Alamelu G. Bharadwaj
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (A.G.B.); (E.K.)
- Departments of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 1X5, Canada
| | - Emma Kempster
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (A.G.B.); (E.K.)
| | - David M. Waisman
- Departments of Pathology, Dalhousie University, Halifax, NS B3H 1X5, Canada; (A.G.B.); (E.K.)
- Departments of Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS B3H 1X5, Canada
- Correspondence: ; Tel.: +1-(902)-494-1803; Fax: +1-(902)-494-1355
| |
Collapse
|
3
|
Mallya K, Gautam SK, Aithal A, Batra SK, Jain M. Modeling pancreatic cancer in mice for experimental therapeutics. Biochim Biophys Acta Rev Cancer 2021; 1876:188554. [PMID: 33945847 DOI: 10.1016/j.bbcan.2021.188554] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy.
Collapse
Affiliation(s)
- Kavita Mallya
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
4
|
Huang J, Agoston AT, Guo P, Moses MA. A Rationally Designed ICAM1 Antibody Drug Conjugate for Pancreatic Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002852. [PMID: 33344137 PMCID: PMC7740099 DOI: 10.1002/advs.202002852] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Indexed: 05/09/2023]
Abstract
Outcomes for pancreatic cancer (PC) patients remain strikingly poor with a 5-year survival of less than 8% due to the lack of effective treatment modalities. Here, a novel precision medicine approach for PC treatment is developed, which is composed of a rationally designed tumor-targeting ICAM1 antibody-drug conjugate (ADC) with optimized chemical linker and cytotoxic payload, complemented with a magnetic resonance imaging (MRI)-based molecular imaging approach to noninvasively evaluate the efficiency of ICAM1 ADC therapy. It is shown that ICAM1 is differentially overexpressed on the surface of human PC cells with restricted expression in normal tissues, enabling ICAM1 antibody to selectively recognize and target PC tumors in vivo. It is further demonstrated that the developed ICAM1 ADC induces potent and durable tumor regression in an orthotopic PC mouse model. To build a precision medicine, an MRI-based molecular imaging approach is developed that noninvasively maps the tumoral ICAM1 expression that can be potentially used to identify ICAM1-overexpressing PC patients. Collectively, this study establishes a strong foundation for the development of a promising ADC to address the critical need in the PC patient care.
Collapse
Affiliation(s)
- Jing Huang
- Vascular Biology ProgramBoston Children's HospitalBostonMA02115USA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Agoston T. Agoston
- Department of PathologyBrigham and Women's HospitalHarvard Medical SchoolBostonMA02115USA
| | - Peng Guo
- Vascular Biology ProgramBoston Children's HospitalBostonMA02115USA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
| | - Marsha A. Moses
- Vascular Biology ProgramBoston Children's HospitalBostonMA02115USA
- Department of SurgeryBoston Children's Hospital and Harvard Medical SchoolBostonMA02115USA
| |
Collapse
|
5
|
Rezaee M, Wang J, Razavi M, Ren G, Zheng F, Hussein A, Ullah M, Thakor AS. A Study Comparing the Effects of Targeted Intra-Arterial and Systemic Chemotherapy in an Orthotopic Mouse Model of Pancreatic Cancer. Sci Rep 2019; 9:15929. [PMID: 31685925 PMCID: PMC6828954 DOI: 10.1038/s41598-019-52490-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
Systemic chemotherapy is the first line treatment for patients with unresectable pancreatic cancer, however, insufficient drug delivery to the pancreas is a major problem resulting in poor outcomes. We evaluated the therapeutic effects of targeted intra-arterial (IA) delivery of gemcitabine directly into the pancreas in an orthotopic mouse model of pancreatic cancer. Nude mice with orthotopic pancreatic tumors were randomly assigned into 3 groups receiving gemcitabine: systemic intravenous (IV) injection (low: 0.3 mg/kg and high: 100 mg/kg) and direct IA injection (0.3 mg/kg). Treatments were administered weekly for 2 weeks. IA treatment resulted in a significantly greater reduction in tumor growth compared to low IV treatment. To achieve a comparable reduction in tumor growth as seen with IA treatment, gemcitabine had to be given IV at over 300x the dose (high IV treatment) which was associated with some toxicity. After 2 weeks, tumor samples from animals treated with IA gemcitabine had significantly lower residual cancer cells, higher cellular necrosis and evidence of increased apoptosis when compared to animals treated with low IV gemcitabine. Our study shows targeted IA injection of gemcitabine directly into the pancreas, via its arterial blood supply, has a superior therapeutic effect in reducing tumor growth compared to the same concentration administered by conventional systemic injection.
Collapse
MESH Headings
- Administration, Intravenous
- Animals
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/therapeutic use
- Cell Line, Tumor
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Administration Schedule
- Female
- Humans
- Infusions, Intra-Arterial
- Male
- Mice
- Mice, Nude
- Neoplasm, Residual
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Transplantation, Heterologous
- Gemcitabine
Collapse
Affiliation(s)
- Melika Rezaee
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
- Chicago Medical School, Rosalind Franklin University, North Chicago, Illinois, 60064, USA
| | - Jing Wang
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mehdi Razavi
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Gang Ren
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Fengyan Zheng
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Ahmed Hussein
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Stanford University School of Medicine, Department of Radiology, Palo Alto, California, 94304, USA.
| |
Collapse
|
6
|
Awasthi N, Mikels-Vigdal AJ, Stefanutti E, Schwarz MA, Monahan S, Smith V, Schwarz RE. Therapeutic efficacy of anti-MMP9 antibody in combination with nab-paclitaxel-based chemotherapy in pre-clinical models of pancreatic cancer. J Cell Mol Med 2019; 23:3878-3887. [PMID: 30941918 PMCID: PMC6533474 DOI: 10.1111/jcmm.14242] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 02/01/2019] [Indexed: 12/22/2022] Open
Abstract
Matrix metalloproteinase 9 (MMP9) is involved in the proteolysis of extracellular proteins and plays a critical role in pancreatic ductal adenocarcinoma (PDAC) progression, invasion and metastasis. The therapeutic potential of an anti‐MMP9 antibody (αMMP9) was evaluated in combination with nab‐paclitaxel (NPT)‐based standard cytotoxic therapy in pre‐clinical models of PDAC. Tumour progression and survival studies were performed in NOD/SCID mice. The mechanistic evaluation involved RNA‐Seq, Luminex, IHC and Immunoblot analyses of tumour samples. Median animal survival compared to controls was significantly increased after 2‐week therapy with NPT (59%), Gem (29%) and NPT+Gem (76%). Addition of αMMP9 antibody exhibited further extension in survival: NPT+αMMP9 (76%), Gem+αMMP9 (47%) and NPT+Gem+αMMP9 (94%). Six‐week maintenance therapy revealed that median animal survival was significantly increased after NPT+Gem (186%) and further improved by the addition of αMMP9 antibody (218%). Qualitative assessment of mice exhibited that αMMP9 therapy led to a reduction in jaundice, bloody ascites and metastatic burden. Anti‐MMP9 antibody increased the levels of tumour‐associated IL‐28 (1.5‐fold) and decreased stromal markers (collagen I, αSMA) and the EMT marker vimentin. Subcutaneous tumours revealed low but detectable levels of MMP9 in all therapy groups but no difference in MMP9 expression. Anti‐MMP9 antibody monotherapy resulted in more gene expression changes in the mouse stroma compared to the human tumour compartment. These findings suggest that anti‐MMP9 antibody can exert specific stroma‐directed effects that could be exploited in combination with currently used cytotoxics to improve clinical PDAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, Indiana.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| | | | | | - Margaret A Schwarz
- Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana.,Department of Pediatrics, Indiana University School of Medicine, South Bend, Indiana
| | - Sheena Monahan
- Department of Surgery, Indiana University School of Medicine, South Bend, Indiana
| | | | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, Indiana.,Harper Cancer Research Institute, University of Notre Dame, South Bend, Indiana
| |
Collapse
|
7
|
Bydoun M, Sterea A, Liptay H, Uzans A, Huang WY, Rodrigues GJ, Weaver ICG, Gu H, Waisman DM. S100A10, a novel biomarker in pancreatic ductal adenocarcinoma. Mol Oncol 2018; 12:1895-1916. [PMID: 30009399 PMCID: PMC6210040 DOI: 10.1002/1878-0261.12356] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/05/2018] [Accepted: 06/27/2018] [Indexed: 01/08/2023] Open
Abstract
Pancreatic cancer is arguably the deadliest cancer type. The efficacy of current therapies is often hindered by the inability to predict patient outcome. As such, the development of tools for early detection and risk prediction is key for improving outcome and quality of life. Here, we introduce the plasminogen receptor S100A10 as a novel predictive biomarker and a driver of pancreatic tumor growth and invasion. We demonstrated that S100A10 mRNA and protein are overexpressed in human pancreatic tumors compared to normal ducts and nonductal stroma. S100A10 mRNA and methylation status were predictive of overall survival and recurrence-free survival across multiple patient cohorts. S100A10 expression was driven by promoter methylation and the oncogene KRAS. S100A10 knockdown reduced surface plasminogen activation, invasiveness, and in vivo growth of pancreatic cancer cell lines. These findings delineate the clinical and functional contribution of S100A10 as a biomarker in pancreatic cancer.
Collapse
Affiliation(s)
- Moamen Bydoun
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andra Sterea
- Department of Physiology and Biophysics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Henry Liptay
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Andrea Uzans
- Dalhousie Medical School, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Weei-Yuarn Huang
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gloria J Rodrigues
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychology and Neuroscience, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada.,Brain Repair Centre, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Hong Gu
- Department of Mathematics and Statistics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - David M Waisman
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada.,Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
8
|
Kuc N, Doermann A, Shirey C, Lee DD, Lowe CW, Awasthi N, Schwarz RE, Stahelin RV, Schwarz MA. Pancreatic ductal adenocarcinoma cell secreted extracellular vesicles containing ceramide-1-phosphate promote pancreatic cancer stem cell motility. Biochem Pharmacol 2018; 156:458-466. [PMID: 30222969 DOI: 10.1016/j.bcp.2018.09.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 09/13/2018] [Indexed: 02/06/2023]
Abstract
The high mortality rate associated with pancreatic ductal adenocarcinoma (PDAC) is in part due to lack of effective therapy for this highly chemoresistant tumor. Cancer stem cells, a subset of cancer cells responsible for tumor initiation and metastasis, are not targeted by conventional cytotoxic agents, which renders the identification of factors that facilitate cancer stem cell activation useful in defining targetable mechanisms. We determined that bioactive sphingolipid induced migration of pancreatic cancer stem cells (PCSC) and signaling was specific to ceramide-1-phosphate (C1P). Furthermore, PDAC cells were identified as a rich source of C1P. Importantly, PDAC cells express the C1P converting enzyme ceramide kinase (CerK), secrete C1P-containing extracellular vesicles that mediate PCSC migration, and when co-injected with PCSC reduce animal survival in a PDAC peritoneal dissemination model. Our findings suggest that PDAC secrete C1P-containing extracellular vesicles as a means of recruiting PCSC to sustain tumor growth therefore making C1P release a mechanism that could facilitate tumor progression.
Collapse
Affiliation(s)
- Norbert Kuc
- Department of Biological Sciences, University of Notre Dame, United States
| | - Allison Doermann
- Department of Biological Sciences, University of Notre Dame, United States
| | - Carolyn Shirey
- Department of Chemistry and Biochemistry, University of Notre Dame, United States
| | - Daniel D Lee
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, United States; Department of Pediatrics, Indiana University School of Medicine, United States
| | - Chinn-Woan Lowe
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, United States
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, United States
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, United States; Department of Goshen Center for Cancer Care, Goshen, IN, United States
| | - Robert V Stahelin
- Department of Chemistry and Biochemistry, University of Notre Dame, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, United States
| | - Margaret A Schwarz
- Department of Chemistry and Biochemistry, University of Notre Dame, United States; Department of Cellular and Integrative Physiology, Indiana University School of Medicine, United States; Department of Pediatrics, Indiana University School of Medicine, United States.
| |
Collapse
|
9
|
A Novel Immunocompetent Mouse Model of Pancreatic Cancer with Robust Stroma: a Valuable Tool for Preclinical Evaluation of New Therapies. J Gastrointest Surg 2016; 20:53-65; discussion 65. [PMID: 26582596 PMCID: PMC5724755 DOI: 10.1007/s11605-015-2985-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 10/10/2015] [Indexed: 01/31/2023]
Abstract
A valid preclinical tumor model should recapitulate the tumor microenvironment. Immune and stromal components are absent in immunodeficient models of pancreatic cancer. While these components are present in genetically engineered models such as Kras(G12D); Trp53(R172H); Pdx-1Cre (KPC), immense variability in development of invasive disease makes them unsuitable for evaluation of novel therapies. We have generated a novel mouse model of pancreatic cancer by implanting tumor fragments from KPC mice into the pancreas of wild type mice. Three-millimeter tumor pieces from KPC mice were implanted into the pancreas of C57BL/6J mice. Four to eight weeks later, tumors were harvested, and stromal and immune components were evaluated. The efficacy of Minnelide, a novel compound which has been shown to be effective against pancreatic cancer in a number of preclinical murine models, was evaluated. In our model, consistent tumor growth and metastases were observed. Tumors demonstrated intense desmoplasia and leukocytic infiltration which was comparable to that in the genetically engineered KPC model and significantly more than that observed in KPC tumor-derived cell line implantation model. Minnelide treatment resulted in a significant decrease in the tumor weight and volume. This novel model demonstrates a consistent growth rate and tumor-associated mortality and recapitulates the tumor microenvironment. This convenient model is a valuable tool to evaluate novel therapies.
Collapse
|
10
|
Awasthi N, Hinz S, Brekken RA, Schwarz MA, Schwarz RE. Nintedanib, a triple angiokinase inhibitor, enhances cytotoxic therapy response in pancreatic cancer. Cancer Lett 2014; 358:59-66. [PMID: 25527450 DOI: 10.1016/j.canlet.2014.12.027] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 02/06/2023]
Abstract
Angiogenesis remains a sensible target for pancreatic ductal adenocarcinoma (PDAC) therapy. VEGF, PDGF, FGF and their receptors are expressed at high levels and correlate with poor prognosis in human PDAC. Nintedanib is a triple angiokinase inhibitor that targets VEGFR1/2/3, FGFR1/2/3 and PDGFRα/β signaling. We investigated the antitumor activity of nintedanib alone or in combination with the cytotoxic agent gemcitabine in experimental PDAC. Nintedanib inhibited proliferation of cells from multiple lineages found in PDAC, with gemcitabine enhancing inhibitory effects. Nintedanib blocked PI3K/MAPK activity and induced apoptosis in vitro and in vivo. In a heterotopic model, net local tumor growth compared to controls (100%) was 60.8 ± 10.5% in the gemcitabine group, -2.1 ± 9.9% after nintedanib therapy and -12.4 ± 16% after gemcitabine plus nintedanib therapy. Effects of therapy on intratumoral proliferation, microvessel density and apoptosis corresponded with tumor growth inhibition data. In a PDAC survival model, median animal survival after gemcitabine, nintedanib and gemcitabine plus nintedanib was 25, 31 and 38 days, respectively, compared to 16 days in controls. The strong antitumor activity of nintedanib in experimental PDAC supports the potential of nintedanib-controlled mechanisms as targets for improved clinical PDAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617; Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390.
| | - Stefan Hinz
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Rolf A Brekken
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, South Bend, IN 46617
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN 46617; Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390; Indiana University Health Goshen Center for Cancer Care, Indiana University School of Medicine, South Bend, IN 46617
| |
Collapse
|
11
|
Awasthi N, Zhang C, Schwarz AM, Hinz S, Schwarz MA, Schwarz RE. Enhancement of nab-paclitaxel antitumor activity through addition of multitargeting antiangiogenic agents in experimental pancreatic cancer. Mol Cancer Ther 2014; 13:1032-43. [PMID: 24608575 DOI: 10.1158/1535-7163.mct-13-0361] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nanoparticle albumin-bound paclitaxel (nab-paclitaxel, NPT) has recently shown efficacy in pancreatic ductal adenocarcinoma (PDAC). Targeting tumor angiogenesis is a sensible combination therapeutic strategy for cancer, including PDAC. We tested the hypothesis that NPT response in PDAC can be enhanced by the mechanistically different antiangiogenic agents bevacizumab (Bev) or sunitinib (Su), despite its inherently increased tumor penetration and drug delivery. Compared with controls (19 days), median animal survival was increased after NPT therapy (32 days, a 68% increase, P = 0.0008); other regimens with enhanced survival were NPT+Bev (38 days, a 100% increase, P = 0.0004), NPT+Su (37 days, a 95% increase, P = 0.0004), and NPT+Bev+Su (49 days, a 158% increase, P = 0.0001) but not bevacizumab, sunitinib, or Bev+Su therapy. Relative to controls (100 ± 22.8), percentage net local tumor growth was 28.2 ± 23.4 with NPT, 55.6 ± 18 (Bev), 38.8 ± 30.2 (Su), 11 ± 7.2 (Bev+Su), 32.8 ± 29.2 (NPT+Bev), 6.6 ± 10.4 (NPT+Su), and 13.8 ± 12.5 (NPT+Bev+Su). Therapeutic effects on intratumoral proliferation, apoptosis, microvessel density, and stromal density corresponded with tumor growth inhibition data. In AsPC-1 PDAC cells, NPT IC(50) was reduced >6-fold by the addition of sunitinib (IC(25)) but not by bevacizumab. In human umbilical vein endothelial cells (HUVEC), NPT IC(50) (82 nmol/L) was decreased to 41 nmol/L by bevacizumab and to 63 nmol/L by sunitinib. In fibroblast WI-38 cells, NPT IC(50) (7.2 μmol/L) was decreased to 7.8 nmol/L by sunitinib, but not by bevacizumab. These findings suggest that the effects of one of the most active cytotoxic agents against PDAC, NPT, can be enhanced with antiangiogenic agents, which clinically could relate to greater responses and improved antitumor results.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Authors' Affiliations: Departments of Surgery and Pediatrics; Indiana University Health Goshen Center for Cancer Care, Indiana University School of Medicine, South Bend, Indiana; Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas; and Department of Gastrointestinopancreatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | | | | | | | | | | |
Collapse
|
12
|
Lee JJ, Huang J, England CG, McNally LR, Frieboes HB. Predictive modeling of in vivo response to gemcitabine in pancreatic cancer. PLoS Comput Biol 2013; 9:e1003231. [PMID: 24068909 PMCID: PMC3777914 DOI: 10.1371/journal.pcbi.1003231] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 08/03/2013] [Indexed: 01/03/2023] Open
Abstract
A clear contradiction exists between cytotoxic in-vitro studies demonstrating effectiveness of Gemcitabine to curtail pancreatic cancer and in-vivo studies failing to show Gemcitabine as an effective treatment. The outcome of chemotherapy in metastatic stages, where surgery is no longer viable, shows a 5-year survival <5%. It is apparent that in-vitro experiments, no matter how well designed, may fail to adequately represent the complex in-vivo microenvironmental and phenotypic characteristics of the cancer, including cell proliferation and apoptosis. We evaluate in-vitro cytotoxic data as an indicator of in-vivo treatment success using a mathematical model of tumor growth based on a dimensionless formulation describing tumor biology. Inputs to the model are obtained under optimal drug exposure conditions in-vitro. The model incorporates heterogeneous cell proliferation and death caused by spatial diffusion gradients of oxygen/nutrients due to inefficient vascularization and abundant stroma, and thus is able to simulate the effect of the microenvironment as a barrier to effective nutrient and drug delivery. Analysis of the mathematical model indicates the pancreatic tumors to be mostly resistant to Gemcitabine treatment in-vivo. The model results are confirmed with experiments in live mice, which indicate uninhibited tumor proliferation and metastasis with Gemcitabine treatment. By extracting mathematical model parameter values for proliferation and death from monolayer in-vitro cytotoxicity experiments with pancreatic cancer cells, and simulating the effects of spatial diffusion, we use the model to predict the drug response in-vivo, beyond what would have been expected from sole consideration of the cancer intrinsic resistance. We conclude that this integrated experimental/computational approach may enhance understanding of pancreatic cancer behavior and its response to various chemotherapies, and, further, that such an approach could predict resistance based on pharmacokinetic measurements with the goal to maximize effective treatment strategies.
Collapse
Affiliation(s)
- James J. Lee
- School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Justin Huang
- School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
| | - Christopher G. England
- Department of Pharmacology/Toxicology, University of Louisville, Louisville, Kentucky, United States of America
| | - Lacey R. McNally
- School of Medicine, University of Louisville, Louisville, Kentucky, United States of America
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail: (LRM); (HBF)
| | - Hermann B. Frieboes
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States of America
- Department of Bioengineering, University of Louisville, Louisville, Kentucky, United States of America
- * E-mail: (LRM); (HBF)
| |
Collapse
|
13
|
Ostapoff KT, Awasthi N, Cenik BK, Hinz S, Dredge K, Schwarz RE, Brekken RA. PG545, an angiogenesis and heparanase inhibitor, reduces primary tumor growth and metastasis in experimental pancreatic cancer. Mol Cancer Ther 2013; 12:1190-201. [PMID: 23696215 DOI: 10.1158/1535-7163.mct-12-1123] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Aggressive tumor progression, metastasis, and resistance to conventional therapies lead to an extremely poor prognosis for pancreatic ductal adenocarcinoma (PDAC). Heparanase, an enzyme expressed by multiple cell types, including tumor cells in the tumor microenvironment, has been implicated in angiogenesis and metastasis, and its expression correlates with decreased overall survival in PDAC. We evaluated the therapeutic potential of PG545, an angiogenesis and heparanase inhibitor, in experimental PDAC. PG545 inhibited the proliferation, migration, and colony formation of pancreatic cancer cells in vitro at pharmacologically relevant concentrations. Heparanase inhibition also reduced the proliferation of fibroblasts but had only modest effects on endothelial cells in vitro. Furthermore, PG545 significantly prolonged animal survival in intraperitoneal and genetic models (mPDAC: LSL-Kras(G12D); Cdkn2a(lox/lox); p48(Cre)) of PDAC. PG545 also inhibited primary tumor growth and metastasis in orthotopic and genetic endpoint studies. Analysis of tumor tissue revealed that PG545 significantly decreased cell proliferation, increased apoptosis, reduced microvessel density, disrupted vascular function, and elevated intratumoral hypoxia. Elevated hypoxia is a known driver of collagen deposition and tumor progression; however, tumors from PG545-treated animals displayed reduced collagen deposition and a greater degree of differentiation compared with control or gemcitabine-treated tumors. These results highlight the potent antitumor activity of PG545 and support the further exploration of heparanase inhibitors as a potential clinical strategy for the treatment of PDAC.
Collapse
Affiliation(s)
- Katherine T Ostapoff
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390, USA.
| | | | | | | | | | | | | |
Collapse
|
14
|
Awasthi N, Zhang C, Hinz S, Schwarz MA, Schwarz RE. Enhancing sorafenib-mediated sensitization to gemcitabine in experimental pancreatic cancer through EMAP II. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2013; 32:12. [PMID: 23497499 PMCID: PMC3618297 DOI: 10.1186/1756-9966-32-12] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Accepted: 02/20/2013] [Indexed: 12/19/2022]
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive human malignancies and tends to be relatively resistant to conventional therapies. Activated Ras oncogene mutations are found in up to 90% of PDAC, leading to activation of the Ras/Raf/MEK/ERK signaling pathway. Sorafenib is a multikinase inhibitor of the Ras/Raf/MEK/ERK pathway and of tumor angiogenesis. Endothelial monocyte activating polypeptide II (EMAP) enhances gemcitabine effects in PDAC. Antitumor activity of sorafenib was evaluated in combination with gemcitabine (Gem) and the antiangiogenic agent EMAP in experimental PDAC. Methods Cell proliferation and protein expression were analyzed by WST-1 assay and Western blotting. Animal survival studies were performed in murine PDAC xenografts. Results Sorafenib decreased phospho-MEK, phospho-ERK1/2, phospho-p70S6K and phospho-4EBP-1 expression in PDAC cells. Sorafenib inhibited in vitro proliferation of all four PDAC cell lines tested. Additive effects on cell proliferation inhibition were observed in the gemcitabine-sorafenib combination in PDAC cells, and in combinations of sorafenib or EMAP with gemcitabine in endothelial (HUVEC) and fibroblast (WI-38) cells. Sorafenib, alone or in combination with gemcitabine and EMAP, induced apoptosis in HUVECs and WI-38 cells as observed via increased expression of cleaved poly (ADP-ribose) polymerase-1 (PARP-1) and caspase-3 proteins. Compared to controls (median survival: 22 days), animal survival increased after Gem therapy (29 days) but not in sorafenib (23 days) or EMAP therapy alone (25 days). Further increases in survival occurred in combination therapy groups Gem+sorafenib (30 days, p=0.004), Gem+EMAP (33 days, p=0.002), and Gem+sorafenib+EMAP (36 days, p=0.004), but not after the sorafenib+EMAP combination (24 days). Conclusions These findings demonstrate that the addition of a polymechanistic antiangiogenic agent such as EMAP can enhance the combination treatment effects of sorafenib and cytotoxic PDAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
15
|
Awasthi N, Zhang C, Ruan W, Schwarz MA, Schwarz RE. BMS-754807, a small-molecule inhibitor of insulin-like growth factor-1 receptor/insulin receptor, enhances gemcitabine response in pancreatic cancer. Mol Cancer Ther 2012; 11:2644-53. [PMID: 23047891 DOI: 10.1158/1535-7163.mct-12-0447] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Gemcitabine has limited clinical benefits in pancreatic ductal adenocarcinoma (PDAC). Insulin-like growth factor (IGF) signaling proteins are frequently overexpressed in PDAC. The therapeutic potential of BMS-754807, a small-molecule inhibitor of IGF-type 1 receptor (IGF-1R) and insulin receptor (IR), and gemcitabine was evaluated in experimental PDAC. Cell proliferation and protein expression were measured by WST-1 assay and immunoblotting. Tumor growth and survival studies were conducted in murine xenografts. PDAC cells expressed phospho-IGF-1R protein. BMS-754807 and gemcitabine inhibited cell proliferation of PDAC cells; the combination of BMS-754807 with gemcitabine had additive effects. Addition of BMS-754807 decreased gemcitabine IC₅₀ from 9.7 μmol/L to 75 nmol/L for AsPC-1, from 3 μmol/L to 70 nmol/L for Panc-1, from 72 to 16 nmol/L for MIA PaCa-2, and from 28 to 16 nmol/L for BxPC-3 cells. BMS-754807 caused a decrease in phospho-IGF-1R and phospho-AKT proteins in AsPC-1 and Panc-1 cells. BMS-754807 and gemcitabine caused an increase in PARP-1 and caspase-3 cleavage. Net tumor growth inhibition in BMS-754807, gemcitabine, and BMS-754807+gemcitabine groups was 59%, 35%, and 94% as compared with controls. Effects of therapy on intratumoral proliferation and apoptosis corresponded with tumor growth inhibition data. BMS-754807 also caused a decrease in phospho-IGF-1R and phospho-AKT in tumor tissue lysates. Median animal survival (controls: 21 days) with BMS-754807 was 27 days (P = 0.03), with gemcitabine 28 days (P = 0.05), and in the BMS-754807+gemcitabine combination group, 41 days (P = 0.007). The strong antitumor activity of BMS-754807 in experimental PDAC supports the potential of BMS-754807-induced mechanisms for clinical PDAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Simmons Comprehensive Cancer Center, The University of Texas Southern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | |
Collapse
|
16
|
Awasthi N, Yen PL, Schwarz MA, Schwarz RE. The efficacy of a novel, dual PI3K/mTOR inhibitor NVP-BEZ235 to enhance chemotherapy and antiangiogenic response in pancreatic cancer. J Cell Biochem 2012; 113:784-91. [PMID: 22020918 DOI: 10.1002/jcb.23405] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Gemcitabine has limited clinical benefits for pancreatic ductal adenocarcinoma (PDAC). The phosphatidylinositol-3-kinase (PI3K)/AKT and mammalian target of rapamycin (mTOR) signaling pathways are frequently dysregulated in PDAC. We investigated the effects of NVP-BEZ235, a novel dual PI3K/mTOR inhibitor, in combination with gemcitabine and endothelial monocyte activating polypeptide II (EMAP) in experimental PDAC. Cell proliferation and protein expression were analyzed by WST-1 assay and Western blotting. Animal survival experiments were performed in murine xenografts. BEZ235 caused a decrease in phospho-AKT and phospho-mTOR expression in PDAC (AsPC-1), endothelial (HUVECs), and fibroblast (WI-38) cells. BEZ235 inhibited in vitro proliferation of all four PDAC cell lines tested. Additive effects on proliferation inhibition were observed in the BEZ235-gemcitabine combination in PDAC cells and in combination of BEZ235 or EMAP with gemcitabine in HUVECs and WI-38 cells. BEZ235, alone or in combination with gemcitabine and EMAP, induced apoptosis in AsPC-1, HUVECs, and WI-38 cells as observed by increased expression of cleaved poly (ADP-ribose) polymerase-1 (PARP-1) and caspase-3 proteins. Compared to controls (median survival: 16 days), animal survival increased after BEZ235 and EMAP therapy alone (both 21 days) and gemcitabine monotherapy (28 days). Further increases in survival occurred in combination therapy groups BEZ235 + gemcitabine (30 days, P = 0.007), BEZ235 + EMAP (27 days, P = 0.02), gemcitabine + EMAP (31 days, P = 0.001), and BEZ235 + gemcitabine + EMAP (33 days, P = 0.004). BEZ235 has experimental PDAC antitumor activity in vitro and in vivo that is further enhanced by combination of gemcitabine and EMAP. These findings demonstrate advantages of combination therapy strategies targeting multiple pathways in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | | | | | | |
Collapse
|
17
|
Evaluation of poly-mechanistic antiangiogenic combinations to enhance cytotoxic therapy response in pancreatic cancer. PLoS One 2012; 7:e38477. [PMID: 22723862 PMCID: PMC3377661 DOI: 10.1371/journal.pone.0038477] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Accepted: 05/09/2012] [Indexed: 12/17/2022] Open
Abstract
Gemcitabine (Gem) has limited clinical benefits in pancreatic ductal adenocarcinoma (PDAC). The present study investigated combinations of gemcitabine with antiangiogenic agents of various mechanisms for PDAC, including bevacizumab (Bev), sunitinib (Su) and EMAP II. Cell proliferation and protein expression were analyzed by WST-1 assay and Western blotting. In vivo experiments were performed via murine xenografts. Inhibition of in vitro proliferation of AsPC-1 PDAC cells by gemcitabine (10 µM), bevacizumab (1 mg/ml), sunitinib (10 µM) and EMAP (10 µM) was 35, 22, 81 and 6 percent; combination of gemcitabine with bevacizumab, sunitinib or EMAP had no additive effects. In endothelial HUVECs, gemcitabine, bevacizumab, sunitinib and EMAP caused 70, 41, 86 and 67 percent inhibition, while combination of gemcitabine with bevacizumab, sunitinib or EMAP had additive effects. In WI-38 fibroblasts, gemcitabine, bevacizumab, sunitinib and EMAP caused 79, 58, 80 and 29 percent inhibition, with additive effects in combination as well. Net in vivo tumor growth inhibition in gemcitabine, bevacizumab, sunitinib and EMAP monotherapy was 43, 38, 94 and 46 percent; dual combinations of Gem+Bev, Gem+Su and Gem+EMAP led to 69, 99 and 64 percent inhibition. Combinations of more than one antiangiogenic agent with gemcitabine were generally more effective but not superior to Gem+Su. Intratumoral proliferation, apoptosis and microvessel density findings correlated with tumor growth inhibition data. Median animal survival was increased by gemcitabine (26 days) but not by bevacizumab, sunitinib or EMAP monotherapy compared to controls (19 days). Gemcitabine combinations with bevacizumab, sunitinib or EMAP improved survival to similar extent (36 or 37 days). Combinations of gemcitabine with Bev+EMAP (43 days) or with Bev+Su+EMAP (46 days) led to the maximum survival benefit observed. Combination of antiangiogenic agents improves gemcitabine response, with sunitinib inducing the strongest effect. These findings demonstrate advantages of combining multi-targeting agents with standard gemcitabine therapy for PDAC.
Collapse
|
18
|
Awasthi N, Schwarz MA, Schwarz RE. Antitumour activity of sunitinib in combination with gemcitabine in experimental pancreatic cancer. HPB (Oxford) 2011; 13:597-604. [PMID: 21843259 PMCID: PMC3183443 DOI: 10.1111/j.1477-2574.2011.00333.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Gemcitabine (Gem) has limited clinical benefits in pancreatic ductal adenocarcinoma (PDAC). Sunitinib (Su) is a novel, multi-target receptor tyrosine kinase inhibitor that has antitumour activities. This study tested the benefits of combined gemcitabine and sunitinib in PDAC. METHODS Cell viability and protein expression were evaluated by WST-1 assay and Western blotting. Tumour growth and survival experiments were performed in murine xenografts. RESULTS In PDAC cells, Gem, Su and Su + Gem, respectively, caused 28%, 22% and 48% inhibition in proliferation at 100 nM. In endothelial cells, Gem, Su and Su + Gem, respectively, caused 49%, 32% and 72% inhibition in proliferation. In fibroblasts, Gem, Su and Su + Gem, respectively, caused 65%, 14% and 79% inhibition in proliferation. Su increased apoptosis, as evidenced by the cleavage of caspase-3 and PARP-1 proteins. Net tumour growth compared with controls in the Gem, Su and Su + Gem groups was 57%, 6% and 1%, respectively. Intratumoral proliferative activity was reduced by 33%, 82% and 75% in the Gem, Su and Su + Gem groups, respectively, compared with controls. Median survival in the control, Su, Gem and Su + Gem groups was 16, 21, 24 and 30 days, respectively (P=0.007). CONCLUSIONS These findings support a combination approach using multi-target antiangiogenic agents such as sunitinib with standard gemcitabine therapy in the treatment of PDAC.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical CenterDallas, TX, USA,Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Margaret A Schwarz
- Department of Pediatrics, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical CenterDallas, TX, USA
| | - Roderich E Schwarz
- Division of Surgical Oncology, Department of Surgery, University of Texas Southwestern Medical CenterDallas, TX, USA,Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
19
|
Awasthi N, Kirane A, Schwarz MA, Toombs JE, Brekken RA, Schwarz RE. Smac mimetic-derived augmentation of chemotherapeutic response in experimental pancreatic cancer. BMC Cancer 2011; 11:15. [PMID: 21226944 PMCID: PMC3034706 DOI: 10.1186/1471-2407-11-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 01/12/2011] [Indexed: 01/09/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) is highly resistant to conventional chemotherapy, in part due to the overexpression of inhibitors of apoptosis proteins (IAPs). Smac is an endogenous IAP-antagonist, which renders synthetic Smac mimetics attractive anticancer agents. We evaluated the benefits of combining a Smac mimetic, JP1201 (JP), with conventional chemotherapy agents used for PDAC management. Methods Cell viability assays and protein expression analysis were performed using WST-1 reagent and Western blotting, respectively. Apoptosis was detected by annexin V/propidium iodide staining. In vivo tumor growth and survival studies were performed in murine PDAC xenografts. Results JP and gemcitabine (Gem) inhibited PDAC cell proliferation with additive effects in combination. The percentage of early apoptotic cells in controls, JP, Gem and JP + Gem was 17%, 26%, 26% and 38%, respectively. JP-induced apoptosis was accompanied by PARP-1 cleavage. Similar additive anti-proliferative effects were seen for combinations of JP with doxorubicin (Dox) and docetaxel (DT). The JP + Gem combination caused a 30% decrease in tumor size in vivo compared to controls. Median animal survival was improved significantly in mice treated with JP + Gem (38 d) compared to controls (22 d), JP (28 d) or Gem (32 d) (p = 0.01). Animal survival was also improved with JP + DT treatment (32 d) compared to controls (16 d), JP (21 d) or DT alone (27 d). Conclusions These results warrant further exploration of strategies that promote chemotherapy-induced apoptosis of tumors and highlight the potential of Smac mimetics in clinical PDAC therapy.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Department of Surgery, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | |
Collapse
|
20
|
Awasthi N, Schwarz MA, Schwarz RE. Enhancing cytotoxic agent activity in experimental pancreatic cancer through EMAP II combination therapy. Cancer Chemother Pharmacol 2010; 68:571-82. [PMID: 21110024 DOI: 10.1007/s00280-010-1514-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Accepted: 10/26/2010] [Indexed: 12/11/2022]
Abstract
PURPOSE Gemcitabine has limited benefits as single agent or in combination for pancreatic ductal adenocarcinoma (PDAC). Endothelial monocyte-activating polypeptide II (EMAP) enhances gemcitabine effects in PDAC. We evaluated the combination effects of EMAP, doxorubicin, and docetaxel in experimental PDAC. METHODS Cell proliferation, protein expression, and apoptosis were analyzed by WST-1 assay, Western blotting, and FACS analysis. Tumor growth and survival experiments were performed in murine xenografts. RESULTS PDAC cell proliferation in vitro was not affected by EMAP, compared to a small inhibition through doxorubicin, docetaxel, and gemcitabine. EMAP addition to these agents did not increase the antiproliferative effects. In endothelial cells, EMAP, doxorubicin, docetaxel, and gemcitabine all had antiproliferative effects. Addition of EMAP to these cytotoxic agents had additive effects. In PDAC cells, no agent induced measurable apoptosis, whereas in endothelial cells, all agents either alone or in combination did. Doxorubicin, docetaxel, gemcitabine, and EMAP all decreased tumor growth. EMAP addition increased inhibitory effects of docetaxel and gemcitabine, but not of doxorubicin. However, compared to controls (median survival: 17 days), EMAP (14 days) had no survival benefit, while docetaxel (29 days), gemcitabine (25 days), and docetaxel followed by gemcitabine sequence (37 days) extended animal survival. Addition of EMAP to docetaxel (35 days), gemcitabine (28 days), and docetaxel gemcitabine sequence (41 days) extended the survival. Doxorubicin effects were not enhanced by EMAP. CONCLUSIONS The antiendothelial combination therapy benefit through EMAP is not limited to gemcitabine and may facilitate the development of more effective alternative cytotoxic therapy strategies against PDAC.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390-8548, USA
| | | | | |
Collapse
|
21
|
Tseng WW, Winer D, Kenkel JA, Choi O, Shain AH, Pollack JR, French R, Lowy AM, Engleman EG. Development of an orthotopic model of invasive pancreatic cancer in an immunocompetent murine host. Clin Cancer Res 2010; 16:3684-95. [PMID: 20534740 DOI: 10.1158/1078-0432.ccr-09-2384] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE The most common preclinical models of pancreatic adenocarcinoma utilize human cells or tissues that are xenografted into immunodeficient hosts. Several immunocompetent, genetically engineered mouse models of pancreatic cancer exist; however, tumor latency and disease progression in these models are highly variable. We sought to develop an immunocompetent, orthotopic mouse model of pancreatic cancer with rapid and predictable growth kinetics. EXPERIMENTAL DESIGN Cell lines with epithelial morphology were derived from liver metastases obtained from Kras(G12D/+);LSL-Trp53(R172H/+);Pdx-1-Cre mice. Tumor cells were implanted in the pancreas of immunocompetent, histocompatible B6/129 mice, and the mice were monitored for disease progression. Relevant tissues were harvested for histologic, genomic, and immunophenotypic analysis. RESULTS All mice developed pancreatic tumors by two weeks. Invasive disease and liver metastases were noted by six to eight weeks. Histologic examination of tumors showed cytokeratin-19-positive adenocarcinoma with regions of desmoplasia. Genomic analysis revealed broad chromosomal changes along with focal gains and losses. Pancreatic tumors were infiltrated with dendritic cells, myeloid-derived suppressor cells, macrophages, and T lymphocytes. Survival was decreased in RAG(-/-) mice, which are deficient in T cells, suggesting that an adaptive immune response alters the course of disease in wild-type mice. CONCLUSIONS We have developed a rapid, predictable orthotopic model of pancreatic adenocarcinoma in immunocompetent mice that mimics human pancreatic cancer with regard to genetic mutations, histologic appearance, and pattern of disease progression. This model highlights both the complexity and relevance of the immune response to invasive pancreatic cancer and may be useful for the preclinical evaluation of new therapeutic agents.
Collapse
Affiliation(s)
- William W Tseng
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
In vivo bioluminescent imaging of irradiated orthotopic pancreatic cancer xenografts in nonobese diabetic-severe combined immunodeficient mice: a novel method for targeting and assaying efficacy of ionizing radiation. Transl Oncol 2010; 3:153-9. [PMID: 20563256 DOI: 10.1593/tlo.09184] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2009] [Revised: 12/17/2009] [Accepted: 12/29/2009] [Indexed: 01/23/2023] Open
Abstract
Adenocarcinoma of the pancreas is a lethal malignancy, and better models to study tumor behavior in vivo are needed for the development ofmore effective therapeutics. Ionizing radiation is a treatment modality that is commonly used in the clinical setting, in particular, for locally confined disease; however, good model systems to study the effect of ionizing radiation in orthotopic tumors have not been established. In an attempt to create clinically relevant models for studying treatments directed against pancreatic cancer, we have defined a methodology to measure the effect of varying doses of radiation in established human pancreatic cancer orthotopic xenografts using two different pancreatic cancer cell lines (Panc-1 and BXPC3) infected with a lentiviral vector expressing CMV promoter-driven luciferase to allow bioluminescence imaging of live animals in real time. Quantifiable photon emission from luciferase signaling in vivo correlated well with actual tumor growth. Bioluminescence imaging of the established pancreatic xenografts was used to direct delivery of radiation to the orthotopic tumors and minimize off-target adverse effects. Growth delay was observed with schedules in the range of 7.5 Gy in five fractions to 10 Gy in four fractions, whereas doses 3 Gy or higher produced toxic adverse effects. In conclusion, we describe a model in which the effects of ionizing radiation, alone or in combination with other therapeutics, in orthotopic xenografts, can be studied.
Collapse
|
23
|
Samkoe KS, Chen A, Rizvi I, O'Hara JA, Hoopes PJ, Pereira SP, Hasan T, Pogue BW. Imaging tumor variation in response to photodynamic therapy in pancreatic cancer xenograft models. Int J Radiat Oncol Biol Phys 2010; 76:251-9. [PMID: 20005458 PMCID: PMC2902770 DOI: 10.1016/j.ijrobp.2009.08.041] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 07/20/2009] [Accepted: 08/06/2009] [Indexed: 01/06/2023]
Abstract
PURPOSE A treatment monitoring study investigated the differential effects of orthotopic pancreatic cancer models in response to interstitial photodynamic therapy (PDT), and the validity of using magnetic resonance imaging as a surrogate measure of response was assessed. METHODS AND MATERIALS Different orthotopic pancreatic cancer xenograft models (AsPC-1 and Panc-1) were used to represent the range of pathophysiology observed in human beings. Identical dose escalation studies (10, 20, and 40J/cm) using interstitial verteporfin PDT were performed, and magnetic resonance imaging with T2-weighted and T1-weighted contrast were used to monitor the total tumor volume and the vascular perfusion volume, respectively. RESULTS There was a significant amount of necrosis in the slower-growing Panc-1 tumor using high light dose, although complete necrosis was not observed. Lower doses were required for the same level of tumor kill in the faster-growing AsPC-1 cell line. CONCLUSIONS The tumor growth rate and vascular pattern of the tumor affect the optimal PDT treatment regimen, with faster-growing tumors being relatively easier to treat. This highlights the fact that therapy in human beings shows a heterogeneous range of outcomes, and suggests a need for careful individualized treatment outcomes assessment in clinical work.
Collapse
|
24
|
Schwarz RE, Awasthi N, Konduri S, Cafasso D, Schwarz MA. EMAP II-Based Antiangiogenic-Antiendothelial In Vivo Combination Therapy of Pancreatic Cancer. Ann Surg Oncol 2009; 17:1442-52. [DOI: 10.1245/s10434-009-0879-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Indexed: 01/04/2023]
|
25
|
Awasthi N, Schwarz MA, Schwarz RE. Proteasome inhibition enhances antitumour effects of gemcitabine in experimental pancreatic cancer. HPB (Oxford) 2009; 11:600-5. [PMID: 20495713 PMCID: PMC2785956 DOI: 10.1111/j.1477-2574.2009.00109.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 06/16/2009] [Indexed: 12/12/2022]
Abstract
BACKGROUND The clinical benefit of gemcitabine, the standard systemic therapy of pancreatic cancer (PaCa), remains modest as a result of high chemoresistance. The proteasome inhibitor bortezomib has antitumour activity against PaCa in vitro and in vivo. We examined the antitumour activity of combination PaCa therapy with bortezomib and gemcitabine. METHODS Cell proliferation assays were performed using WST-1 reagent. Protein expression was determined by Western blotting. Efficacy of bortezomib and/or gemcitabine was tested in vivo in a survival study. RESULTS Bortezomib at 10 microM caused 29% and 72% inhibition in AsPC-1 PaCa cell proliferation at 48 and 96 h incubation, respectively. Bortezomib was even more active against PaCa cell lines Panc-1 and MiaPaCa, with 80% inhibition of proliferation at 48 h. The combination of bortezomib and gemcitabine inhibited AsPC-1 proliferation more effectively compared with each single agent alone. Poly(ADP-ribose) polymerase (PARP) cleavage, an apoptotic indicator, reached 6.6-, 2- and 8.5-fold over controls for bortezomib, gemcitabine and the combination. The median survival was 31 (controls and bortezomib), 40 (gemcitabine) and 48 days (combination), respectively (P < 0.002). CONCLUSIONS Bortezomib and gemcitabine demonstrate additive antitumour activity in vitro and in an experimental PaCa model, indicating the potential for clinical PaCa benefits of additional multiagent therapies that will be based upon the bortezomib and gemcitabine combination.
Collapse
Affiliation(s)
- Niranjan Awasthi
- Division of Surgical Oncology, Department of Surgery, UT Southwestern Medical CenterDallas, TX, USA
| | | | - Roderich E Schwarz
- Division of Surgical Oncology, Department of Surgery, UT Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
26
|
An antiendothelial combination therapy strategy to increase survival in experimental pancreatic cancer. Surgery 2009; 146:241-9. [DOI: 10.1016/j.surg.2009.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2009] [Accepted: 04/17/2009] [Indexed: 12/17/2022]
|
27
|
Lebedeva IV, Su ZZ, Vozhilla N, Chatman L, Sarkar D, Dent P, Athar M, Fisher PB. Chemoprevention by perillyl alcohol coupled with viral gene therapy reduces pancreatic cancer pathogenesis. Mol Cancer Ther 2008; 7:2042-50. [PMID: 18645014 DOI: 10.1158/1535-7163.mct-08-0245] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer is one of the deadliest of cancers. Even with aggressive therapy, the 5-year survival rate is <5%, mandating development of more effective treatments. Melanoma differentiation-associated gene-7/interleukin-24 (mda-7/IL-24) shows potent antitumor activity against most cancers displaying safety with significant clinical efficacy. However, pancreatic cancer cells display inherent resistance to mda-7/IL-24 that is the result of a "protein translational block" of mda-7/IL-24 mRNA in these tumor cells. We now show that a dietary supplement perillyl alcohol (POH) has significant chemopreventive effects for pancreatic cancer and, when coupled with adenovirus-mediated mda-7/IL-24 gene therapy (Ad.mda-7), effectively eliminates s.c. and i.p. xenografts of human pancreatic cancer cells in nude mice, promoting enhanced survival. The combination of POH and Ad.mda-7 efficiently abrogates the mda-7/IL-24 protein translational block, resulting in MDA-7/IL-24 protein production and growth suppression. Of direct translational relevance, clinically achievable concentrations of POH with Ad.mda-7, both of which have been found safe and without toxic effects in human trials, were used. This novel and innovative approach combining a dietary agent and a virally delivered therapeutic cytokine provides a means of both preventing and treating human pancreatic cancer with significant clinical translational potential.
Collapse
Affiliation(s)
- Irina V Lebedeva
- Department of Urology, Herbert Irving Comprehensive Cancer Center, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Hamada K, Ueda N, Ito M, Roth JA, Follen M. The nude rat as an orthotopic model for cervical cancer. Gynecol Oncol 2005; 99:S159-65. [PMID: 16185752 DOI: 10.1016/j.ygyno.2005.07.073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The purposes of this study were to establish intracervical tumors of the nude rat as an orthotopic experimental model for human cervical cancer and to preliminary evaluate the effects of the adenoviral vector, Ad5CMV-p53, on orthotopic cervical tumor size. METHODS Human cervical cancer SiHa and ME-180 cells were injected into the cervix of the nude rat. Four days later, 1 x 10(9) plaque forming units (PFU) of Ad5CMV-p53 were injected into the cervix. The rats were later sacrificed to determine cervical tumor size. RESULTS Eight of ten nude rats developed SiHa cell tumors; all ten nude rats developed ME-180 cell tumors. Four of ten SiHa cell tumors metastasized to the pelvic cavity; no ME-180 cell tumors did. The growth of Ad5CMV-p53-infected cells was greatly suppressed. The ad5CMV-p53 treatment significantly reduced both cell tumor volumes in nude rat cervixes. CONCLUSION The nude rat cervix grows tumors similar to human cervical cancer tumors and makes an excellent experimental model. Transfection of cervical cancer cells with the wild-type p53 gene via Ad5CMV-p53 is a potential therapeutic approach to cervical cancer.
Collapse
Affiliation(s)
- Katsuyuki Hamada
- Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | |
Collapse
|
29
|
Thakur VS, Shankar B, Chatterjee S, Premachandran S, Sainis KB. Role of tumor-derived transforming growth factor-beta1 (TGF-beta1) in site-dependent tumorigenicity of murine ascitic lymphosarcoma. Cancer Immunol Immunother 2005; 54:837-47. [PMID: 15726358 PMCID: PMC11032987 DOI: 10.1007/s00262-004-0656-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2004] [Accepted: 12/07/2004] [Indexed: 02/04/2023]
Abstract
An ascitic lymphosarcoma (LS-A) of Swiss mice that regressed spontaneously on subcutaneous (s.c.) transplantation was investigated for the mechanism of its progressive growth and host mortality on intraperitoneal (i.p.) transplantation. In vitro studies indicated significant inhibition of LS-A proliferation seeded at higher cell density (>10(4)/ml). Culture supernatants of LS-A caused bi-modal growth effects, the early supernatants (24 h) caused stimulation and the late (72 h) supernatants inhibited LS-A proliferation. The 72-h supernatants also suppressed T and B cell response to mitogens in a dose-dependent manner. Pan anti-transforming growth factor-beta antibody abrogated the inhibitory effects of supernatants. The supernatants contained both latent as well as bio-active form of transforming growth factor-beta1 (TGF-beta1) as determined by ELISA. Mice bearing i.p. ascites tumor had elevated serum TGF-beta1, hemoglobulinemia, splenic lymphopenia, impaired response of the T cells to mitogen and reduced expression of transferrin receptor (CD71) on the bone marrow cells. However, mice which rejected s.c. transplants, did not show significant changes in these parameters. Our studies indicated profound influence of site of tumor growth on tumor progression and host immune system mediated by tumor-derived TGF-beta1. It is possible that human tumors which secrete TGF-beta1 may exhibit similar patho-physiological effects in the host depending on the anatomical site of the tumor.
Collapse
MESH Headings
- Anemia/blood
- Anemia/etiology
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, B-Lymphocyte/metabolism
- Ascites
- Cell Proliferation
- Disease Progression
- Hemoglobins/analysis
- Humans
- Injections, Intraperitoneal
- Injections, Subcutaneous
- Lymphoma, Non-Hodgkin/immunology
- Lymphoma, Non-Hodgkin/metabolism
- Lymphoma, Non-Hodgkin/pathology
- Lymphopenia/etiology
- Lymphopenia/metabolism
- Lymphopenia/pathology
- Male
- Mice
- Mitogens/pharmacology
- Receptors, Transferrin
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/metabolism
- Sarcoma, Experimental/pathology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta1
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- V. S. Thakur
- Radiation Biology & Health Sciences Division, Bioscience Group, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085 India
| | - B. Shankar
- Radiation Biology & Health Sciences Division, Bioscience Group, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085 India
| | - S. Chatterjee
- Radiation Biology & Health Sciences Division, Bioscience Group, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085 India
| | - S. Premachandran
- Radiation Biology & Health Sciences Division, Bioscience Group, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085 India
| | - K. B. Sainis
- Radiation Biology & Health Sciences Division, Bioscience Group, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085 India
| |
Collapse
|
30
|
Zagon IS, Jaglowski JR, Verderame MF, Smith JP, Leure-Dupree AE, McLaughlin PJ. Combination chemotherapy with gemcitabine and biotherapy with opioid growth factor (OGF) enhances the growth inhibition of pancreatic adenocarcinoma. Cancer Chemother Pharmacol 2005; 56:510-20. [PMID: 15947928 DOI: 10.1007/s00280-005-1028-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Accepted: 02/07/2005] [Indexed: 01/14/2023]
Abstract
Gemcitabine is the standard of care for advanced pancreatic neoplasia, and exerts its effect through inhibition of DNA synthesis. However, gemcitabine has limited survival benefits. Opioid growth factor (OGF) is an autocrine-produced peptide that interacts with the nuclear receptor, OGFr, to inhibit cell proliferation but is not cytotoxic or apoptotic. The present study was designed to examine whether a combination of chemotherapy with gemcitabine and biotherapy with OGF is more effective than either agent alone in inhibiting pancreatic cancer growth in vitro and in vivo. The combination of OGF (10(-6) M) and gemcitabine (10(-8) M) reduced MIA PaCa-2 cell number from control levels by 46% within 48 h, and resulted in a growth inhibition greater than that of the individual compounds. OGF in combination with 5-fluorouracil also depressed cell growth more than either agent alone. The action of OGF, but not gemcitabine, was mediated by a naloxone-sensitive receptor, and was completely reversible. OGF, but no other endogenous or exogenous opioids, altered pancreatic cancer growth in tissue culture. The combination of OGF and gemcitabine also repressed the growth of another pancreatic cancer cell line, PANC-1. MIA PaCa-2 cells transplanted into athymic mice received 10 mg/kg OGF daily, 120 mg/kg gemcitabine every 3 days; 10 mg/kg OGF daily and 120 mg/kg gemcitabine every 3rd day, or 0.1 ml of sterile saline daily. Tumor incidence, and latency times to tumor appearance, of mice receiving combined therapy with OGF and gemcitabine, were significantly decreased from those of the control, OGF, and gemcitabine groups. Tumor volumes in the OGF, gemcitabine, and OGF/gemcitabine groups were markedly decreased from controls beginning on days 14, 12, and 8, respectively, after tumor cell inoculation. Tumor weight and tumor volume were reduced from control levels by 36-85% in the OGF and/or gemcitabine groups on day 45 (date of termination), and the group of mice exposed to a combination of OGF and gemcitabine had decreases in tumor size of 70% and 63% from the OGF or the gemcitabine alone groups, respectively. This preclinical evidence shows that combined chemotherapy (e.g. gemcitabine) and biotherapy (OGF) provides an enhanced therapeutic benefit for pancreatic cancer.
Collapse
Affiliation(s)
- Ian S Zagon
- Department of Neural and Behavioral Sciences, H109, Milton S. Hershey Medical Center, The Pennsylvania State University, Hershey, 17033, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Tirone TA, Wang XP, Templeton NS, Lee T, Nguyen L, Fisher W, Brunicardi FC. Cell-specific cytotoxicity of human pancreatic adenocarcinoma cells using rat insulin promoter thymidine kinase-directed gene therapy. World J Surg 2004; 28:826-33. [PMID: 15457366 DOI: 10.1007/s00268-004-7291-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The formation of a normal pancreas and the activation of insulin production are, in part, dependent on the expression and activation of the pancreatic duodenal homeobox gene 1 (PDX-1). The expression of PDX-1 also has been detected in various human pancreatic ductal adenocarcinoma (PDA) cell lines. This has made it possible to generate a cancer cell-specific gene expression system to treat human pancreatic cancer. In this study, we have developed a cell-specific cytotoxic model of PDA cells using the expression of herpes simplex virus thymidine kinase (TK) under the control of the rat insulin promoter (RIP-TK). We have shown that the cell-specific cytotoxicity in human PDA cells depends on the presence of PDX-1. Our results also demonstrate that in vivo PDA-specific cytotoxicity can be achieved with RIP-TK using an intraperitoneal liposomal gene delivery method followed by a short period of ganciclovir treatment in severe combined immunodeficient (SCID) mice. Furthermore, PDX-1 protein was found in all six freshly isolated human pancreas cancer specimens and two liver metastasis samples that were group-tested, suggesting the feasibility of using RIP-TK gene therapy in humans. This study may provide an alternative strategy for the future treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Thomas A Tirone
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, 6550 Fannin, Suite 1661, 77030, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Fogar P, Greco E, Basso D, Habeler W, Navaglia F, Zambon CF, Tormen D, Gallo N, Cecchetto A, Plebani M, Pedrazzoli S. Suicide gene therapy with HSV-TK in pancreatic cancer has no effect in vivo in a mouse model. Eur J Surg Oncol 2004; 29:721-30. [PMID: 14602490 DOI: 10.1016/j.ejso.2003.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM To study in vivo whether pancreatic cancer tumour growth and metastasis can be modified by a gene construct with HSV-TK suicide gene and IL2 co-expression. METHODS Seventy-eight female SCID mice were i.p. inoculated with retrovirally transduced or control MIA PaCa 2, CAPAN-1 and PANC-1 cell lines. The animals were then randomly selected for saline or ganciclovir (GCV) treatment from the second week, for a total of two weeks. RESULTS Most inoculated mice developed tumour nodules and spleen metastases. The liver was colonized by control CAPAN-1 and MIA PaCa 2, but not by PANC-1. Tumours in transduced MIA PaCa 2 cell injected mice were smaller, and in transduced CAPAN-1 injected mice larger, than in control-inoculated mice. There were increased pancreatic and decreased spleen metastases from transduced CAPAN-1, and diminished liver involvement from transduced MIA PaCa 2. No differences were found between mice inoculated with transduced and control PANC-1 cell lines. GCV treatment had no effect on tumour's size or metastases. CONCLUSIONS The HSV-TK suicide gene does not confer GCV sensitivity to pancreatic cancer in this in vivo model. Different pancreatic cancer cell lines cause different growth and metastasis patterns after inoculation in SCID mice, possibly because of variations in their inherent characteristics. The different effects of our vector on cell growth and metastasis may be attributable to the effects of the immunostimulatory cytokine IL2.
Collapse
Affiliation(s)
- P Fogar
- Department of Medical and Surgical Sciences, University of Padua, Clinica Chirurgica IV, Via Giustiniani 2, 35128 Padua, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Katz MH, Takimoto S, Spivack D, Moossa AR, Hoffman RM, Bouvet M. A novel red fluorescent protein orthotopic pancreatic cancer model for the preclinical evaluation of chemotherapeutics. J Surg Res 2003; 113:151-60. [PMID: 12943825 DOI: 10.1016/s0022-4804(03)00234-8] [Citation(s) in RCA: 110] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Realistic models of pancreatic cancer are necessary to develop effective drugs for the disease. More aggressive tumor models enhanced by brighter fluorescent biomarkers to follow the disease in real time would enhance the ability to predict accurately the effect of novel therapeutics on this particularly malignant human cancer. MATERIALS AND METHODS A novel, highly fluorescent, red fluorescent protein (RFP)-expressing pancreatic cancer model was orthotopically established in nude mice. The MIA-PaCa-2 human pancreatic cancer cell line was transduced with RFP and grown subcutaneously. Fluorescent tumor fragments were then surgically transplanted onto the nude mouse pancreas. Groups treated with intraperitoneal gemcitabine or intravenous irinotecan were sequentially imaged to compare, in real time, the antimetastatic and antitumor effects of these agents compared with untreated controls. RESULTS Rapid tumor growth and widespread metastases developed in untreated mice within 2 weeks, leading to a median survival of 21 days. In contrast, significant tumor growth suppression and consequent increase in survival (32.5 days, P = 0.009) were achieved with CPT-11. Gemcitabine highly improved survival (72 days, P = 0.004) by inducing transient tumor regression over the first 3 weeks. However, at this time, growth and dissemination occurred despite continued treatment, suggesting the development of tumor resistance. The antimetastatic efficacy of each drug was followed noninvasively in real time by imaging the RFP-expressing tumor and metastases, and was confirmed by fluorescent open imaging of autopsy specimens. CONCLUSIONS This highly metastatic model reliably simulates the aggressive course of human pancreatic cancer. Noninvasive, sequential imaging permits quantification of tumor growth and dissemination and, thereby, real time evaluation of therapeutic efficacy. These features make this model an ideal, preclinical system with which to study novel therapeutics for pancreatic cancer.
Collapse
Affiliation(s)
- Matthew H Katz
- Department of Surgery, University of California at San Diego, San Diego, California 92161, USA
| | | | | | | | | | | |
Collapse
|
34
|
Keyes KA, Mann L, Teicher B, Alvarez E. Site-dependent angiogenic cytokine production in human tumor xenografts. Cytokine 2003; 21:98-104. [PMID: 12670449 DOI: 10.1016/s1043-4666(03)00015-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Tumor microenvironment plays a critical role in tumor growth, angiogenesis, and metastasis. Differences in site of tumor implantation result in differences in tumor growth, metastasis, as well as response to chemotherapy. We hypothesized that tumor-induced angiogenic growth factor production into the plasma will also be influenced by site of tumor implantation. We evaluated the site-dependent production of angiogenic growth factors in the plasma of tumor bearing animals at two different sites of implantation. Plasma levels of tumor necrosis factor-alpha (TNF-alpha), basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (VEGF) were evaluated in nude mice bearing A2780, SKOV-3, or OVCAR-3 human ovarian tumors, as well as Panc-1, AsPC-1, or BxPC-3 human pancreatic tumors grown as subcutaneous (SC) xenografts or in the intraperitoneal (IP) cavity. Plasma VEGF and bFGF levels produced by two ovarian tumor lines and two pancreatic tumor lines were substantially higher when the tumors were implanted in the IP cavity than in the SC space. These studies indicated that the site of tumor implantation was an important determinant in the production of plasma VEGF and bFGF levels. As more and more anti-angiogenic agents are developed, the need for appropriate animal models becomes apparent. These results suggest the demand for an appropriate model for the in vivo evaluation of anti-angiogenesis.
Collapse
Affiliation(s)
- Kristan A Keyes
- Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, IN 46285, USA.
| | | | | | | |
Collapse
|
35
|
Greco E, Fogar P, Basso D, Stefani AL, Navaglia F, Zambon CF, Mazza S, Gallo N, Piva MG, Scarpa A, Pedrazzoli S, Plebani M. Retrovirus-mediated herpes simplex virus thymidine kinase gene transfer in pancreatic cancer cell lines: an incomplete antitumor effect. Pancreas 2002; 25:e21-9. [PMID: 12142751 DOI: 10.1097/00006676-200208000-00020] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The transfer of drug-susceptible (suicide) genes to tumor cells by retroviral or adenoviral vectors is a novel approach to the treatment of human tumors. AIMS To ascertain the antitumor effect of retroviral transduction of the pancreatic cancer cell lines MIA PaCa 2, CAPAN-1, PANC1, and PSN1 with the herpes simplex virus thymidine kinase (HSV-TK) gene. METHODOLOGY The vector carried a neoselectable marker gene, the human interleukin-2 gene, an internal ribosome entry coding site, and the region coding HSV-TK. RESULTS Twenty micromoles or less of ganciclovir did not modify nontransduced TK- cell growth, whereas > or =100 micromol completely inhibited TK- cell growth, indicating that this dosage is cytotoxic per se. The 4 TK- and the 4 transduced cell lines were treated daily with 0.001, 0.01, 0.1, 1, 10, and 20 micromol of ganciclovir for 13 days. CAPAN-1 cell growth was completely inhibited by 0.1 micromol of ganciclovir; higher doses were required to kill PANC1 (10 micromol) and PSN1 (20 micromol). MIA PaCa 2 cell growth decreased following a 20-micromol ganciclovir dosing. The bystander effect was great in the CAPAN-1 cell line and moderate in PANC1; no bystander effect was recorded in MIA PaCa 2 and PSN1 cell lines. CONCLUSION Gene therapy with HSV-TK for pancreatic cancer seems effective in only a limited number of tumor-derived cell lines, and this limits its application in vivo.
Collapse
Affiliation(s)
- Eliana Greco
- Department of Laboratory Medicine, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ridwelski K, Meyer F, Hribaschek A, Kasper U, Lippert H. Intraoperative and early postoperative chemotherapy into the abdominal cavity using gemcitabine may prevent postoperative occurence of peritoneal carcinomatosis. J Surg Oncol 2002; 79:10-6. [PMID: 11754372 DOI: 10.1002/jso.10000] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND AND OBJECTIVES Malignant tumors of various abdominal organs are blamed to be the cause of death in approximately 40% of cases per year in Western Europe. At the time of death, about half of them show peritoneal carcinomatosis which is considered an unfavorable prognostic factor. In case of peritoneal carcinomatosis, there is no curative treatment available for any of the tumors in the abdominal cavity. The aim of this experimental study was to evaluate the preventive potential of intra-operative or early postoperative local administration of gemcitabine into the abdominal cavity against peritoneal carcinomatosis. METHODS Peritoneal carcinomatosis was induced in male WAG rats (n = 18) by transfer of 5 x 10(6) cells of the adenocarcinoma cell line CC-531 via minilaparotomy carried out in all rats under general anesthesia. Thirty days after cell transfer, rats were sacrificed and peritoneal carcinomatosis was confirmed using histologic investigation in specimens obtained from the peritoneal site. Extension of this carcinomatosis was quantified by (i) counting tumor nodes per square centimeter (mean of several counts) and (ii) determining tumor weight (weight of the greater omentum plus the resected mesentery). Rats were subdivided into three groups (n = 6 per group): group 1 (controls); group 2, simultaneously with tumor cell transfer 24 mg/kg of gemcitabine were administered into the abdominal cavity; group 3, postoperative intraperitoneal irrigation with 24 mg/kg of gemcitabine was achieved via a previously implanted port-a-cath on days(d) 15, 21, and 27. RESULTS On the 30th postoperative day, all six animals of the control group showed extensive tumor growth at the peritoneum and greater omentum, indicating carcinomatosis. In contrast, none of the rats of group 2 revealed any traces of intraperitoneal tumor growth. While all rats of group 3 demonstrated intraperitoneal tumor growth, but early postoperative treatment with gemcitabine, however, resulted in a significant reduction of the number of tumor nodes and tumor weight in comparison with group 1 (controls). CONCLUSIONS Immediate, i.e., simultaneous intraoperative application of a cytostatic agent such as gemcitabine into the abdominal cavity may completely prevent the generation of peritoneal carcinomatosis from occuring tumor cells, whereas early postoperative locoregional chemotherapy into the abdominal cavity can only reduce the severity of the peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Karsten Ridwelski
- Division of Oncosurgery/Oncology, Department of Surgery, University Hospital, Otto von Guericke University, Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
37
|
Alves F, Contag S, Missbach M, Kaspareit J, Nebendahl K, Borchers U, Heidrich B, Streich R, Hiddemann W. An orthotopic model of ductal adenocarcinoma of the pancreas in severe combined immunodeficient mice representing all steps of the metastatic cascade. Pancreas 2001; 23:227-35. [PMID: 11590317 DOI: 10.1097/00006676-200110000-00002] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Clinically relevant animal models are needed to evaluate new therapeutic strategies against pancreatic adenocarcinoma, which is almost incurable by established treatment. AIMS To establish and characterize a metastatic orthotopic transplant model for pancreatic ductal adenocarcinoma in severe combined immunodeficient (SCID) mice. METHODOLOGY Human pancreatic ductal carcinoma cells, PancTu 1, were implanted either subcutaneously or orthotopically into the pancreas. RESULTS After 4 weeks, orthotopic transplantation resulted in an extensive local tumor growth of an undifferentiated ductal adenocarcinoma with slight to moderate desmoplastic reaction. The tumor growth and spread resembled the situation in humans, including invasion into adjacent organs causing biliary and stomach obstruction. In addition, tumor metastases to regional lymph nodes of the pancreas, lung, liver, mesentery, and diaphragm, and attached to the kidneys, spleen, and reproductive organs were observed. In contrast, no invasion or metastases could be demonstrated by subcutaneous implanted PancTu I cells. Using immunohistochemical analysis, even single human tumor cells could be detected in blood vessels and metastatic organs, providing evidence that the orthotopic transplant model appropriately reflects the entire process of the metastatic cascade. CONCLUSION This cancer model in SCID mice appears to be a powerful tool to investigate the identity of metastasis-associated genes and to evaluate preclinically the potency of novel antimetastatic agents in ductal adenocarcinoma of the pancreas.
Collapse
Affiliation(s)
- F Alves
- Department of Hematology and Oncology, Georg-August-University, Göttingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fosh BG, Finch JG, Anthony AA, Texler M, Maddern GJ. Electrolytic ablation of the rat pancreas: a feasibility trial. BMC Gastroenterol 2001; 1:9. [PMID: 11570977 PMCID: PMC56592 DOI: 10.1186/1471-230x-1-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2001] [Accepted: 09/06/2001] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pancreatic cancer is a biologically aggressive disease with less than 20% of patients suitable for a "curative" surgical resection. This, combined with the poor 5-year survival indicates that effective palliative methods for symptom relief are required. Currently there are no ablative techniques to treat pancreatic cancer in clinical use. Tissue electrolysis is the delivery of a direct current between an anode and cathode to induce localised necrosis. Electrolysis has been shown to be safe and reliable in producing hepatic tissue and tumour ablation in animal models and in a limited number of patients. This study investigates the feasibility of using electrolysis to produce localised pancreatic necrosis in a healthy rat model. METHOD Ten rats were studied in total. Eight rats were treated with variable "doses" of coulombs, and the systemic and local effects were assessed; 2 rats were used as controls. RESULTS Seven rats tolerated the procedure well without morbidity or mortality, and one died immediately post procedure. One control rat died on induction of anaesthesia. Serum amylase and glucose were not significantly affected. CONCLUSION Electrolysis in the rat pancreas produced localised necrosis and appears both safe, and reproducible. This novel technique could offer significant advantages for patients with unresectable pancreatic tumours. The next stage of the study is to assess pancreatic electrolysis in a pig model, prior to human pilot studies.
Collapse
Affiliation(s)
- Beverley G Fosh
- University of Adelaide, Department Of Surgery, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Jonathon Guy Finch
- University of Adelaide, Department Of Surgery, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Adrian A Anthony
- University of Adelaide, Department Of Surgery, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Michael Texler
- The Department Of Pathology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Guy J Maddern
- University of Adelaide, Department Of Surgery, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| |
Collapse
|
39
|
Hori T, Yamashita Y, Ohira M, Matsumura Y, Muguruma K, Hirakawa K. A novel orthotopic implantation model of human esophageal carcinoma in nude rats: CD44H mediates cancer cell invasion in vitro and in vivo. Int J Cancer 2001; 92:489-96. [PMID: 11304682 DOI: 10.1002/ijc.1234] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
A new orthotopic esophageal cancer model was developed by implanting fragments of xenografts of T.T human esophageal squamous carcinoma cells into the cervical esophagus of athymic rats. The rats had symptoms analogous to the human clinical course such as respiratory distress, dysphagia, vomiting of blood, or Horner syndrome, followed by death resulting from suffocation. Microscopic metastases of lymph node were observed around the tumor in 3 of 18 rats. A new cell line (T.T-1) was established from these metastases. Flow cytometry showed that T.T-1 and T.T parental cells had nearly the same surface levels of beta1-integrin, alpha2-integrin, alpha3-integrin and E-cadherin, and no expression of CD44v3, CD44v6 and alpha5-integrin. T.T-1 cells had a higher level of CD44H, however, and a greater binding efficiency to the extracellular matrix components; laminin, type IV collagen, hyaluronic acid, and fibronectin than T.T cells. Anti-CD44H antibody significantly decreased the binding efficiency of T.T-1 cells. T.T-1 cells were also significantly more invasive than T.T cells through all the extracellular matrix components except hyaluronic acid. After orthotopic implantation histological examination showed that T.T-1 tumors invaded beyond the esophageal mucosa and tracheal muscle layer and obstructed the esophagus and trachea. No invasion was observed with T.T tumors. Rats with T.T-1 or T.T tumors survived an average of 32.0 and 50.7 days, respectively (p < 0.01). In addition T.T-1 tumors expressed higher levels of CD44H mRNA than T.T tumors. In summary, our newly developed orthotopic implantation model is a valid model of esophageal cancer because it followed the same clinical course experienced by humans. Moreover, using cells derived from this model, we were able to demonstrate that CD44H is involved in esophageal cancer cell invasion.
Collapse
Affiliation(s)
- T Hori
- First Department of Surgery, Osaka City University Medical School, Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
40
|
Hoge CW, Sethabutr O, Bodhidatta L, Echeverria P, Robertson DC, Morris JG, Schwarz MA, Schwarz RE. Use of a synthetic oligonucleotide probe to detect strains of non-serovar O1 Vibrio cholerae carrying the gene for heat-stable enterotoxin (NAG-ST). Carcinogenesis 1990; 34:2361-9. [PMID: 2380369 DOI: 10.1093/carcin/bgt227] [Citation(s) in RCA: 103] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
A synthetic oligonucleotide probe was developed to identify the gene for the heat-stable enterotoxin (NAG-ST) of non-serovar O1 Vibrio cholerae. Of 103 non-O1 V. cholerae isolates from Thailand, 31 isolates from Mexico, and 47 isolates from patients in the United States, only 7 (all from Thailand) hybridized with the probe. Probe-positive strains produced significantly higher fluid accumulations in infant mice than probe-negative strains.
Collapse
Affiliation(s)
- C W Hoge
- Department of Medicine, University of Maryland School of Medicine, Baltimore 21201
| | | | | | | | | | | | | | | |
Collapse
|