1
|
Leung PY, Chen W, Sari AN, Sitaram P, Wu PK, Tsai S, Park JI. Erlotinib combination with a mitochondria-targeted ubiquinone effectively suppresses pancreatic cancer cell survival. World J Gastroenterol 2024; 30:714-727. [PMID: 38515951 PMCID: PMC10950623 DOI: 10.3748/wjg.v30.i7.714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/13/2023] [Accepted: 01/17/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND Pancreatic cancer is a leading cause of cancer-related deaths. Increased activity of the epidermal growth factor receptor (EGFR) is often observed in pancreatic cancer, and the small molecule EGFR inhibitor erlotinib has been approved for pancreatic cancer therapy by the food and drug administration. Nevertheless, erlotinib alone is ineffective and should be combined with other drugs to improve therapeutic outcomes. We previously showed that certain receptor tyrosine kinase inhibitors can increase mitochondrial membrane potential (Δψm), facilitate tumor cell uptake of Δψm-sensitive agents, disrupt mitochondrial homeostasis, and subsequently trigger tumor cell death. Erlotinib has not been tested for this effect. AIM To determine whether erlotinib can elevate Δψm and increase tumor cell uptake of Δψm-sensitive agents, subsequently triggering tumor cell death. METHODS Δψm-sensitive fluorescent dye was used to determine how erlotinib affects Δψm in pancreatic adenocarcinoma (PDAC) cell lines. The viability of conventional and patient-derived primary PDAC cell lines in 2D- and 3D cultures was measured after treating cells sequentially with erlotinib and mitochondria-targeted ubiquinone (MitoQ), a Δψm-sensitive MitoQ. The synergy between erlotinib and MitoQ was then analyzed using SynergyFinder 2.0. The preclinical efficacy of the two-drug combination was determined using immune-compromised nude mice bearing PDAC cell line xenografts. RESULTS Erlotinib elevated Δψm in PDAC cells, facilitating tumor cell uptake and mitochondrial enrichment of Δψm-sensitive agents. MitoQ triggered caspase-dependent apoptosis in PDAC cells in culture if used at high doses, while erlotinib pretreatment potentiated low doses of MitoQ. SynergyFinder suggested that these drugs synergistically induced tumor cell lethality. Consistent with in vitro data, erlotinib and MitoQ combination suppressed human PDAC cell line xenografts in mice more effectively than single treatments of each agent. CONCLUSION Our findings suggest that a combination of erlotinib and MitoQ has the potential to suppress pancreatic tumor cell viability effectively.
Collapse
Affiliation(s)
- Pui-Yin Leung
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Anissa N Sari
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Poojitha Sitaram
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Pui-Kei Wu
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Susan Tsai
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| |
Collapse
|
2
|
Kaumaya PTP. B-cell epitope peptide cancer vaccines: a new paradigm for combination immunotherapies with novel checkpoint peptide vaccine. Future Oncol 2020; 16:1767-1791. [PMID: 32564612 PMCID: PMC7426751 DOI: 10.2217/fon-2020-0224] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/26/2020] [Indexed: 12/22/2022] Open
Abstract
In light of the numerous US FDA-approved humanized monoclonal antibodies (mAbs) for cancer immunotherapy, it is surprising that the advancement of B-cell epitope vaccines designed to elicit a natural humoral polyclonal antibody response has not gained traction in the immune-oncology landscape. Passive immunotherapy with humanized mAbs (Trastuzumab [Herceptin®]; Pertuzumab [Perjeta®]) has provided clinical benefit to breast cancer patients, albeit with significant shortcomings including toxicity problems and resistance, high costs, sophisticated therapeutic regimen and long half-life. The role of B-cell humoral immunity in cancer is under appreciated and underdeveloped. We have advanced the idea of active immunotherapy with chimeric B-cell epitope peptides incorporating a 'promiscuous' T-cell epitope that elicits a polyclonal antibody response, which provides safe, cost-effective therapeutic advantage over mAbs. We have created a portfolio of validated B-cell peptide epitopes against multiple receptor tyrosine kinases (HER-1, HER-3, IGF-1R and VEGF). We have successfully translated two HER-2 combination B-cell peptide vaccines in Phase I and II clinical trials. We have recently developed an effective novel PD-1 vaccine. In this article, I will review our approaches and strategies that focus on B-cell epitope cancer vaccines.
Collapse
Affiliation(s)
- Pravin TP Kaumaya
- Department of Obstetrics & Gynecology, College of Medicine, Wexner Medical Center, The James Cancer Hospital & Solove Research Institute, The Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
3
|
Yao H, Xu Z, Li C, Tse MK, Tong Z, Zhu G. Synthesis and Cytotoxic Study of a Platinum(IV) Anticancer Prodrug with Selectivity toward Luteinizing Hormone-Releasing Hormone (LHRH) Receptor-Positive Cancer Cells. Inorg Chem 2019; 58:11076-11084. [PMID: 31393117 DOI: 10.1021/acs.inorgchem.9b01583] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Platinum drugs including cisplatin are widely used in clinics to treat various types of cancer. However, the lack of cancer-cell selectivity is one of the major problems that lead to side effects in normal tissues. Luteinizing hormone-releasing hormone (LHRH) receptors are overexpressed in many types of cancer cells but rarely presented in normal cells, making LHRH receptor a good candidate for cancer targeting. In this study, we report the synthesis and cytotoxic study of a novel platinum(IV) anticancer prodrug functionalized with LHRH peptide. This LHRH-platinum(IV) conjugate is highly soluble in water and quite stable in a PBS buffer. Cytotoxic study reveals that the prodrug selectively targets LHRH receptor-positive cancer cell lines with the cytotoxicities 5-8 times higher than those in LHRH receptor-negative cell lines. In addition, the introduction of LHRH peptide enhances the cellular accumulation in a manner of receptor-mediated endocytosis. Moreover, the LHRH-platinum(IV) prodrug is proved to kill cancer cells by binding to the genomic DNA, inducing apoptosis, and arresting the cell cycle at the G2/M phase. In summary, we report a novel LHRH-platinum(IV) anticancer prodrug having largely improved selectivity toward LHRH receptor-positive cancer cells, relative to cisplatin.
Collapse
Affiliation(s)
- Houzong Yao
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Zoufeng Xu
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Cai Li
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| | - Man-Kit Tse
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China
| | - Zixuan Tong
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China
| | - Guangyu Zhu
- Department of Chemistry , City University of Hong Kong , 83 Tat Chee Avenue , Hong Kong SAR 999077 , People's Republic of China.,City University of Hong Kong Shenzhen Research Institute , Shenzhen 518057 , People's Republic of China
| |
Collapse
|
4
|
Calderon LE, Keeling JK, Rollins J, Black CA, Collins K, Arnold N, Vance DE, Ndinguri MW. Pt-Mal-LHRH, a Newly Synthesized Compound Attenuating Breast Cancer Tumor Growth and Metastasis by Targeting Overexpression of the LHRH Receptor. Bioconjug Chem 2016; 28:461-470. [PMID: 27997127 DOI: 10.1021/acs.bioconjchem.6b00610] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A new targeting chemotherapeutic agent, Pt-Mal-LHRH, was synthesized by linking activated cisplatin to luteinizing hormone releasing hormone (LHRH). The compound's efficacy and selectivity toward 4T1 breast cancer cells were evaluated. Carboplatin was selected as the comparative platinum complex, since the Pt-Mal-LHRH malonate linker chelates platinum in a similar manner to carboplatin. Breast cancer and normal cell viability were analyzed by an MTT assay comparing Pt-Mal-LHRH with carboplatin. Cells were also treated with either Pt-Mal-LHRH or carboplatin to evaluate platinum uptake by ICP-MS and cell migration using an in vitro scratch-migration assay. Tumor volume and metastasis were evaluated using an in vivo 4T1 mouse tumor model. Mice were administered Pt-Mal-LHRH (carboplatin molar equivalent dosage) through ip injection and compared to those treated with carboplatin (5 (mg/kg)/week), no treatment, and LHRH plus carboplatin (unbound) controls. An MTT assay showed a reduction in cell viability (p < 0.01) in 4T1 and MDA-MB-231 breast cancer cells treated with Pt-Mal-LHRH compared to carboplatin. Pt-Mal-LHRH was confirmed to be cytotoxic by flow cytometry using a propidium iodide stain. Pt-Mal-LHRH displayed a 20-fold increase in 4T1 cellular uptake compared to carboplatin. There was a decrease (p < 0.0001) in 4T1 cell viability compared to 3T3 normal fibroblast cells. Treatment with Pt-Mal-LHRH also resulted in a significant decrease in cell-migration compared to carboplatin. In vivo testing found a significant reduction in tumor volume (p < 0.05) and metastatic tumor colonization in the lungs with Pt-Mal-LHRH compared to carboplatin. There was a slight decrease in lung weight and no difference in liver weight between treatment groups. Together, our data indicate that Pt-Mal-LHRH is a more potent and selective chemotherapeutic agent than untargeted carboplatin.
Collapse
Affiliation(s)
- Lindsay E Calderon
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Jonathan K Keeling
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Joseph Rollins
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Carrie A Black
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Kendall Collins
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Nova Arnold
- Department of Biology, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Diane E Vance
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| | - Margaret W Ndinguri
- Department of Chemistry, Eastern Kentucky University , Richmond, Kentucky 40475, United States
| |
Collapse
|
5
|
Liu B, Jia Y, Ma J, Wu S, Jiang H, Cao Y, Sun X, Yin X, Yan S, Shang M, Mao A. Tumor-associated macrophage-derived CCL20 enhances the growth and metastasis of pancreatic cancer. Acta Biochim Biophys Sin (Shanghai) 2016; 48:1067-1074. [PMID: 27797715 DOI: 10.1093/abbs/gmw101] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/18/2016] [Accepted: 08/01/2016] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is an aggressive malignancy with a high metastatic potential that results in a high mortality rate worldwide. Although macrophages have the potential to kill tumor cells and elicit immune responses against tumors, there is evidence that tumor-associated macrophages (TAMs) promote tumor progression and suppress T-cell responses. CC-chemokine ligand 20 (CCL20) and its unique receptor CC-chemokine receptor 6 (CCR6) are exploited by cancer cells for migration and metastasis and play important roles in the development and progression of cancer. Recent studies have shown that the expression of CCL20 is upregulated in pancreatic cancer; however, the mechanism of action of CCL20 remains to be fully elucidated. In this study, the aberrant expression of CCL20 in TAMs of pancreatic cancer tissue, including metastatic pancreatic cancer tissue, was detected. CCL20 expression was considerably higher in macrophages than in pancreatic cancer cell lines, particularly in interleukin-4-treated (M2) macrophages. Using Boyden chamber assays of pancreatic cancer cells, we found that CCL20 secreted by M2 macrophages promoted the migration, epithelial-mesenchymal transition, and invasion of pancreatic cancer cells. RNA interference results showed that CCR6 is a receptor for CCL20 in pancreatic cancer cells, mediating the increased invasive properties of these cells promoted by CCL20. Using a mouse model, we confirmed the roles of CCR6/CCL20 in promoting pancreatic cancer growth and liver metastasis in vivo Our findings provide insight into the important role of macrophage-secreted CCL20 in pancreatic cancer and implicate CCR6/CCL20 as potential therapeutic targets.
Collapse
Affiliation(s)
- Bingyan Liu
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Yiping Jia
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Jun Ma
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Shaoqiu Wu
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Haosheng Jiang
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Yan Cao
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Xianjun Sun
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Xiang Yin
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Shuo Yan
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Mingyi Shang
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Aiwu Mao
- Department of Interventional Radiology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| |
Collapse
|
6
|
Mahran RI, Hagras MM, Sun D, Brenner DE. Bringing Curcumin to the Clinic in Cancer Prevention: a Review of Strategies to Enhance Bioavailability and Efficacy. AAPS JOURNAL 2016; 19:54-81. [DOI: 10.1208/s12248-016-0003-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/04/2016] [Indexed: 01/28/2023]
|
7
|
The Prognostic and Predictive Role of Epidermal Growth Factor Receptor in Surgical Resected Pancreatic Cancer. Int J Mol Sci 2016; 17:ijms17071090. [PMID: 27399694 PMCID: PMC4964466 DOI: 10.3390/ijms17071090] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 06/28/2016] [Accepted: 07/01/2016] [Indexed: 12/28/2022] Open
Abstract
The data regarding the prognostic significance of EGFR (epidermal growth factor receptor) expression and adjuvant therapy in patients with resected pancreatic cancer are insufficient. We retrospectively investigated EGFR status in 357 resected PDAC (pancreatic duct adenocarcinoma) patients using tissue immunohistochemistry and validated the possible role of EGFR expression in predicting prognosis. The analysis was based on excluding the multiple confounding parameters. A negative association was found between overall EGFR status and postoperative survival (p = 0.986). Remarkably, adjuvant chemotherapy and radiotherapy were significantly associated with favorable postoperative survival, which prolonged median overall survival (OS) for 5.8 and 10.2 months (p = 0.009 and p = 0.006, respectively). Kaplan-Meier analysis showed that adjuvant chemotherapy correlated with an obvious survival benefit in the EGFR-positive subgroup rather than in the EGFR-negative subgroup. In the subgroup analyses, chemotherapy was highly associated with increased postoperative survival in the EGFR-negative subgroup (p = 0.002), and radiotherapy had a significant survival benefit in the EGFR-positive subgroup (p = 0.029). This study demonstrated that EGFR expression is not correlated with outcome in resected pancreatic cancer patients. Adjuvant chemotherapy and radiotherapy were significantly associated with improved survival in contrary EGFR expressing subgroup. Further studies of EGFR as a potential target for pancreatic cancer treatment are warranted.
Collapse
|
8
|
Egeli U, Ak S, Cecener G, Tunca B, Tezcan G, Sevinc ED, Kaya E, Dundar HZ, Sarkut P, Ozen Y, Balcin O, Evrensel T, Yerci O, Ugras N. Impact of 3'UTR variation patterns of the KRAS gene on the aggressiveness of pancreatobiliary tumors with the KRAS G13D mutation in a Turkish population. Pancreatology 2016; 16:677-86. [PMID: 27256640 DOI: 10.1016/j.pan.2016.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Several studies have demonstrated the importance of mutations in codons 12, 13 and 61 and variations in the 3' untranslated region (3'UTR) of the KRAS gene, frequently observed genetic events in the progression of pancreatobiliary tumors (PBT). However, limited data exist on the clinical effect of these alterations. The aim of the current study was to clarify the frequency of relevant alterations of the 3'UTR regions of the KRAS gene and the effect of KRAS 3'UTR polymorphisms on the prognosis of patients with codon 12, 13 and 61 mutations in a Turkish population with PBT. METHODS Codons 12, 13, and 61 and 3'UTRs of the KRAS gene were screened by single-strand conformation polymorphism (SSCP) analysis and DNA sequencing in 43 patients and 10 controls. Chi-squared and independent sample T tests were used to evaluate the results of the mutation analysis and clinical features of the patients. RESULTS We defined the c.38G > A (rs112445441, p.G13D) (39.54%) mutation and two 3'UTR variations, c.*4066delA (rs560890523) (23.26%) and c.*4065_*4066delAA (rs57698689) (6.98%), in the KRAS gene of Turkish patients. There was a statistically significant relationship between the c.*4066delA (rs560890523) and c.*4065_*4066delAA (rs57698689) variations and invasion and lymph node metastasis status of the patients (p < 0.001). Compared to patients with c.38G > A (rs112445441, p.G13D), patients with c.*4066delA (rs560890523) and c.38G > A (rs112445441, p.G13D) presented more aggressive tumors with highly invasive features. The present study contributes findings regarding the clinical effects of KRAS alterations in PBT. Based on our study, further investigations are required.
Collapse
Affiliation(s)
- Unal Egeli
- Department of Medical Biology, Medical Faculty, Uludag University, Bursa, Turkey.
| | - Secil Ak
- Department of Medical Biology, Medical Faculty, Uludag University, Bursa, Turkey
| | - Gulsah Cecener
- Department of Medical Biology, Medical Faculty, Uludag University, Bursa, Turkey
| | - Berrin Tunca
- Department of Medical Biology, Medical Faculty, Uludag University, Bursa, Turkey
| | - Gulcin Tezcan
- Department of Medical Biology, Medical Faculty, Uludag University, Bursa, Turkey
| | | | - Ekrem Kaya
- Department of General Surgery, Medical Faculty, Uludag University, Bursa, Turkey
| | - Halit Ziya Dundar
- Department of General Surgery, Medical Faculty, Uludag University, Bursa, Turkey
| | - Pinar Sarkut
- Department of General Surgery, Medical Faculty, Uludag University, Bursa, Turkey
| | - Yilmaz Ozen
- Department of General Surgery, Medical Faculty, Uludag University, Bursa, Turkey
| | - Ozkan Balcin
- Department of General Surgery, Medical Faculty, Uludag University, Bursa, Turkey
| | - Turkkan Evrensel
- Department of Medical Oncology, Medical Faculty, Uludag University, Bursa, Turkey
| | - Omer Yerci
- Department of Pathology, Medical Faculty, Uludag University, Bursa, Turkey
| | - Nesrin Ugras
- Department of Pathology, Medical Faculty, Uludag University, Bursa, Turkey
| |
Collapse
|
9
|
Chandrasegaram MD, Chiam SC, Chen JW, Khalid A, Mittinty ML, Neo EL, Tan CP, Dolan PM, Brooke-Smith ME, Kanhere H, Worthley CS. Distribution and pathological features of pancreatic, ampullary, biliary and duodenal cancers resected with pancreaticoduodenectomy. World J Surg Oncol 2015; 13:85. [PMID: 25890023 PMCID: PMC4348158 DOI: 10.1186/s12957-015-0498-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 02/01/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pancreatic cancer (PC) has the worst survival of all periampullary cancers. This may relate to histopathological differences between pancreatic cancers and other periampullary cancers. Our aim was to examine the distribution and histopathologic features of pancreatic, ampullary, biliary and duodenal cancers resected with a pancreaticoduodenectomy (PD) and to examine local trends of periampullary cancers resected with a PD. METHODS A retrospective review of PD between January 2000 and December 2012 at a public metropolitan database was performed. The institutional ethics committee approved this study. RESULTS There were 142 PDs during the study period, of which 70 cases were pre-2010 and 72 post-2010, corresponding to a recent increase in the number of cases. Of the 142 cases, 116 were for periampullary cancers. There were also proportionately more PD for PC (26/60, 43% pre-2010 vs 39/56, 70% post-2010, P = 0.005). There were 65/116 (56%) pancreatic, 29/116 (25%), ampullary, 17/116 (15%) biliary and 5/116 (4%) duodenal cancers. Nodal involvement occurred more frequently in PC (78%) compared to ampullary (59%), biliary (47%) and duodenal cancers (20%), P = 0.002. Perineural invasion was also more frequent in PC (74%) compared to ampullary (34%), biliary (59%) and duodenal cancers (20%), P = 0.002. Microvascular invasion was seen in 57% pancreatic, 38% ampullary, 41% biliary and 20% duodenal cancers, P = 0.222. Overall, clear margins (R0) were achieved in fewer PC 41/65 (63%) compared to ampullary 27/29 (93%; P = 0.003) and biliary cancers 16/17 (94%; P = 0.014). CONCLUSIONS This study highlights that almost half of PD was performed for cancers other than PC, mainly ampullary and biliary cancers. The volume of PD has increased in recent years with an increased proportion being for PC. PC had higher rates of nodal and perineural invasion compared to ampullary, biliary and duodenal cancers.
Collapse
Affiliation(s)
- Manju D Chandrasegaram
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia. .,Division of Surgery, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia.
| | - Su C Chiam
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia.
| | - John W Chen
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia. .,Flinders Medical Centre, Flinders Drive, Bedford Park, Adelaide, SA, 5042, Australia. .,Flinders University, Sturt Rd, Bedford Park, Adelaide, SA, 5042, Australia.
| | - Aisha Khalid
- Flinders Medical Centre, Flinders Drive, Bedford Park, Adelaide, SA, 5042, Australia.
| | - Murthy L Mittinty
- School of Population Health, University of Adelaide, 178 North Terrace, Adelaide, SA, 5005, Australia.
| | - Eu L Neo
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia. .,Flinders Medical Centre, Flinders Drive, Bedford Park, Adelaide, SA, 5042, Australia.
| | - Chuan P Tan
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia.
| | - Paul M Dolan
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia.
| | - Mark E Brooke-Smith
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia. .,Flinders Medical Centre, Flinders Drive, Bedford Park, Adelaide, SA, 5042, Australia. .,Flinders University, Sturt Rd, Bedford Park, Adelaide, SA, 5042, Australia.
| | - Harsh Kanhere
- Division of Surgery, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia. .,HPB Surgery Unit, Queen Elizabeth Hospital, 28 Woodville Road, Adelaide, SA, 5011, Australia.
| | - Chris S Worthley
- HPB Surgery Unit, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia.
| |
Collapse
|
10
|
Thakur A, Lum LG, Schalk D, Azmi A, Banerjee S, Sarkar FH, Mohommad R. Pan-Bcl-2 inhibitor AT-101 enhances tumor cell killing by EGFR targeted T cells. PLoS One 2012. [PMID: 23185240 PMCID: PMC3501501 DOI: 10.1371/journal.pone.0047520] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is a deadly disease and has the worst prognosis among almost all cancers and is in dire need of new and improved therapeutic strategies. Conditioning of tumor cells with chemotherapeutic drug has been shown to enhance the anti-tumor effects of cancer vaccines and adoptive cell therapy. In this study, we investigated the immunomodulatory effects of pan-Bcl-2 inhibitor AT-101 on pancreatic cancer (PC) cell cytotoxicity by activated T cells (ATC). The effects of AT-101 on cytotoxicity, early apoptosis, and Granzyme B (GrzB) and IFN-γ signaling pathways were evaluated during EGFR bispecific antibody armed ATC (aATC)-mediated killing of L3.6pl and MiaPaCa-2 PC cells pre-sensitized with AT-101. We found that pretreatment of tumor cells with AT-101 enhanced susceptibility of L3.6pl and MiaPaCa-2 tumor cells to ATC and aATC-mediated cytotoxicity, which was in part mediated via enhanced release of cytolytic granule GrzB from ATC and aATC. AT-101-sensitized L3.6pl cells showed up-regulation of IFN-γ-mediated induction in the phosphorylation of Ser727-Stat1 (pS727-Stat1), and IFN-γ induced dephosphorylation of phospho-Tyr705-Stat3 (pY705-Stat3). Priming (conditioning) of PC cells with AT-101 can significantly enhance the anti-tumor activity of EGFRBi armed ATC through increased IFN-γ induced activation of pS727-Stat1 and inhibition of pY705-Stat3 phosphorylation, and resulting in increased ratio of pro-apoptotic to anti-apoptotic proteins. Our results verify enhanced cytotoxicity after a novel chemotherapy conditioning strategy against PC that warrants further in vivo and clinical investigations.
Collapse
Affiliation(s)
- Archana Thakur
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
11
|
Frequencies and prognostic role of KRAS and BRAF mutations in patients with localized pancreatic and ampullary adenocarcinomas. Pancreas 2012; 41:759-66. [PMID: 22699145 DOI: 10.1097/mpa.0b013e31823cd9df] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The frequencies and prognostic role of KRAS and BRAF mutations in patients operated on for pancreatic ductal adenocarcinomas (PDACs) and ampullary adenocarcinomas (A-ACs) are scantily studied. METHODS KRAS and BRAF mutations were analyzed in formalin-fixed, paraffin-embedded tumor samples from primarily chemotherapy-naive patients operated on with radical intentions for PDAC (n = 170) and A-AC (n = 107). RESULTS Eighty percent of PDAC patients had KRAS mutations (codon 12 mutations: 74%) and 67% with A-AC (codon 12 mutations: 54%). BRAF mutations were less common, 16% in PDAC and 12% in A-AC, and no V600E mutations were found. Fourteen percent with PDAC and 7% with A-AC had mutations in both KRAS and BRAF. Multivariate analysis, including KRAS status, stage, and American Society of Anesthesiologists physical status classification system score, demonstrated that KRAS mutations in patients with A-AC were associated with short recurrence-free survival (RFS) (hazard ratio, 2.45; 95% confidence interval, 1.19-5.06; P = 0.015) and overall survival (OS) (1.93, 95% 1.12-3.31; P = 0.018). KRAS mutations in patients with PDAC were not associated with RFS and OS. BRAF mutations were not associated with RFS and OS. CONCLUSIONS KRAS mutations frequencies were high in PDAC and A-AC. KRAS mutations were associated with poor prognosis in patients with A-AC, but not in patients with PDAC.
Collapse
|
12
|
Abstract
OBJECTIVES Pancreatic and periampullary cancers have a high incidence of activating KRAS mutations. The aim of this study was to determine the incidence of KRAS and EGFR mutations in pancreatic and periampullary cancers and their relationship with survival. METHODS One hundred patients undergoing pancreaticoduodenectomy or pancreatic biopsy for cancer were recruited. Samples of formalin-fixed paraffin-embedded or fresh pancreatic tissue were obtained. EGFR was analyzed by DNA sequencing of exons 18 to 21. KRAS was analyzed by pyrosequencing of codons 12, 13, and 61. RESULTS EGFR mutations were found in 2 (2.3%) of 88 assessable cases. One in exon 18 (c.1966C>T, p.Q710X) and 1 in exon 19 (c.2066A>G, p.E734G). A synonymous single-nucleotide polymorphism in exon 20 (c.2361G>A, p.Q787) was identified in 57 (67.8%) of 84 patients studied. Twenty-eight (41.2%) of 68 cases harbored a point mutation in KRAS codon 12 (26 cases) and codon 61 (2 cases). The overall median survival was 308 days (range, 7-2623 days). The presence of KRAS point mutations did not significantly alter median survival time (22.8 vs 28.1 months, P = 0.88). CONCLUSIONS EGFR somatic mutations are rare in pancreatobiliary malignancies. KRAS mutations are less common than previous reports and do not correlate with survival.
Collapse
|
13
|
Molecular biology of pancreatic ductal adenocarcinoma progression: aberrant activation of developmental pathways. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 97:41-78. [PMID: 21074729 DOI: 10.1016/b978-0-12-385233-5.00002-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Embryonic development marks a period of peak tissue growth and morphogenesis in the mammalian lifecycle. Many of the pathways that underlie cell proliferation and movement are relatively quiescent in adult animals but become reactivated during carcinogenesis. This phenomenon has been particularly well documented in pancreatic cancer, where detailed genetic studies and a robust mouse model have permitted investigators to test the role of various developmental signals in cancer progression. In this chapter, we review current knowledge regarding the signaling pathways that act during pancreatic development and the evidence that the reactivation of developmentally important signals is critical for the pathogenesis of this treatment-refractory malignancy.
Collapse
|
14
|
Aggarwal S, Ndinguri MW, Solipuram R, Wakamatsu N, Hammer RP, Ingram D, Hansel W. [DLys(6)]-luteinizing hormone releasing hormone-curcumin conjugate inhibits pancreatic cancer cell growth in vitro and in vivo. Int J Cancer 2011; 129:1611-23. [PMID: 21484797 DOI: 10.1002/ijc.26132] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Accepted: 03/30/2011] [Indexed: 12/30/2022]
Abstract
Pancreatic ductal adenocarcinomas are invariably lethal, and developing effective treatments that have minimal side effects is a challenge. Previous studies from our laboratory have shown that conjugates of cell membrane disrupting lytic peptides and luteinizing hormone releasing hormone (LHRH) target and destroy human prostate and breast cancer cells in xenografts in the nude mouse model (Hansel et al., Mol Cell Endocrinol 2007;260-262:183-9; Hansel et al., Mol Cell Endocrinol 2007;269:26-33), which express LHRH receptors. The objectives of our study were to synthesize a bioconjugate of LHRH analog ([DLys(6)]-LHRH) and a dietary microchemical (curcumin) and test the hypothesis that [DLys(6)]-LHRH-curcumin targets and inhibits pancreatic cancer cell growth in vitro and in vivo. In in vitro studies, we determined by confocal microscopy, flow cytometry analysis and reverse transcriptase-polymerase chain reaction that MIAPaCa-2, Panc-1 and BxPC-3 pancreatic cancer cell lines express LHRH receptors. [DLys(6)]-LHRH-curcumin inhibited cell proliferation of pancreatic cancer cell lines and induced apoptotic cell death (p < 0.05). Apoptosis was induced by cleavage of polyadenosine-5'-diphosphate-ribose-polymerase and caspase-3. The activity of [DLys(6)]-LHRH-curcumin was equal to free curcumin at equimolar concentrations in vitro. Unlike curcumin itself, the [DLys(6)]-LHRH-curcumin conjugate is water soluble which allows its intravenous administration. In two in vivo studies, [DLys(6)]-LHRH-curcumin given intravenously caused a significant (p < 0.01) reduction in tumor weights and volumes, and free curcumin given by gavage at an equal dose failed to cause a significant reduction in tumor weights and volumes in the nude mouse pancreatic cancer model. [DLys(6)]-LHRH-curcumin treatment enhanced apoptosis compared to [DLys(6)]-LHRH and vehicle-treated controls in tumor tissue. In conclusion, [DLys(6)]-LHRH-curcumin may be useful in treating pancreatic cancer.
Collapse
Affiliation(s)
- S Aggarwal
- William Hansel Cancer Prevention Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Epidermal growth factor receptor in pancreatic cancer. Cancers (Basel) 2011; 3:1513-26. [PMID: 24212772 PMCID: PMC3757375 DOI: 10.3390/cancers3021513] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2011] [Revised: 02/28/2011] [Accepted: 03/11/2011] [Indexed: 12/27/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer related death. The difficulty in detecting pancreatic cancer at an early stage, aggressiveness and the lack of effective therapy all contribute to the high mortality. Epidermal growth factor receptor (EGFR) is a transmembrane glycoprotein, which is expressed in normal human tissues. It is a member of the tyrosine kinase family of growth factors receptors and is encoded by proto-oncogenes. Several studies have demonstrated that EGFR is over-expressed in pancreatic cancer. Over-expression correlates with more advanced disease, poor survival and the presence of metastases. Therefore, inhibition of the EGFR signaling pathway is an attractive therapeutic target. Although several combinations of EGFR inhibitors with chemotherapy demonstrate inhibition of tumor-induced angiogenesis, tumor cell apoptosis and regression in xenograft models, these benefits remain to be confirmed. Multimodality treatment incorporating EGFR-inhibition is emerging as a novel strategy in the treatment of pancreatic cancer.
Collapse
|
16
|
Lambe S, Cantwell N, Islam F, Horvath K, Jefferson AL. Perceptions, knowledge, incentives, and barriers of brain donation among African American elders enrolled in an Alzheimer's research program. THE GERONTOLOGIST 2010; 51:28-38. [PMID: 20679141 DOI: 10.1093/geront/gnq063] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE To learn about African American older adults' knowledge and perceptions of brain donation, factors that relate to participating or not participating in a brain donation research program, and methods to increase African American brain donation commitment rates in the context of an Alzheimer's disease (AD) research program. DESIGN AND METHODS African American older adults (n = 15) from the Boston University Alzheimer's Disease Core Center participant research registry enrolled in 1 of 2 focus groups of 90 min about brain donation. Seven participants were selected for a third follow-up focus group. RESULTS Focus group transcripts were analyzed using consensual qualitative research methods, and 8 overarching themes emerged: (a) perceptions of and misconceptions about brain donation procedures, (b) racial minorities in medical research, (c) racial disparities and discrimination in medical settings, (d) influence of religion and spirituality, (e) family perceptions of and involvement in donation, (f) family history of disease and desire to find a cure, (g) prior exposure to medical and research settings, and (h) culturally sensitive approaches to brain donation. IMPLICATIONS Culturally relevant educational protocols need to be created for use with African American older adults. These protocols should include information about brain donation procedures, rates of AD among Black elders, and potential benefits of donation to Black communities; inclusion of religious figures, family, and peers in donation education and decisions; and methods to address mistrust, including cultural competence trainings for staff.
Collapse
Affiliation(s)
- Susan Lambe
- Alzheimer’s Disease Center and Department of Neurology, Boston University School of Medicine, Boston, Massachusetts 02118-2526, USA
| | | | | | | | | |
Collapse
|
17
|
Shi XH, Liang ZY, Ren XY, Liu TH. Combined silencing of K-ras and Akt2 oncogenes achieves synergistic effects in inhibiting pancreatic cancer cell growth in vitro and in vivo. Cancer Gene Ther 2008; 16:227-36. [PMID: 18949011 DOI: 10.1038/cgt.2008.82] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
18
|
Abstract
Pancreatic cancer is the 4th leading cause of cancer-related death in the United States. The number of diagnoses per year equals the number of deaths per year, making it the deadliest of all malignancies. Modern advances and breakthroughs in molecular oncology have allowed researchers to gain a better understanding of the mechanisms responsible for the pathogenesis of this disease. The transforming growth factor-beta (TGF-beta) pathway is one of the signaling systems that has been identified as a major contributor. TGF-beta plays a paradoxical role as both a tumor suppressor and a tumor promoter in pancreatic cancer. The purpose of this review is to provide the practicing clinician a thorough review of this molecule and its associated signaling partners in the context of its duplicitous role and behavior in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Mark J Truty
- Department of Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
19
|
Liao JD, Adsay NV, Khannani F, Grignon D, Thakur A, Sarkar FH. Histological complexities of pancreatic lesions from transgenic mouse models are consistent with biological and morphological heterogeneity of human pancreatic cancer. Histol Histopathol 2007; 22:661-76. [PMID: 17357096 PMCID: PMC3882316 DOI: 10.14670/hh-22.661] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Although pancreatic cancer is the fourth leading cause of cancer death, it has received much less attention compared to other malignancies. There are several transgenic animal models available for studies of pancreatic carcinogenesis, but most of them do not recapitulate, histologically, human pancreatic cancer. Here we review some detailed molecular complexity of human pancreatic cancer and their reflection in histomorphological complexities of pancreatic lesions developed in various transgenic mouse models with a special concern for studying the effects of chemotherapeutic and chemopreventive agents. These studies usually require a large number of animals that are at the same age and gender and should be either homozygote or heterozygote but not a mixture of both. Only single-transgene models can meet these special requirements, but many currently available models require a mouse to simultaneously bear several transgene alleles. Thus it is imperative to identify new gene promoters or enhancers that are specific for the ductal cells of the pancreas and are highly active in vivo so as to establish new single-transgene models that yield pancreatic ductal adenocarcinomas for chemotherapeutic and chemopreventive studies.
Collapse
Affiliation(s)
- J D Liao
- Department of Pathology, Wayne State University School of Medicine, Karmanos Cancer Institute, Detroit, Michigan 48201, USA
| | | | | | | | | | | |
Collapse
|
20
|
Yeh JJ, Der CJ. Targeting signal transduction in pancreatic cancer treatment. Expert Opin Ther Targets 2007; 11:673-94. [PMID: 17465725 DOI: 10.1517/14728222.11.5.673] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Pancreatic cancer is a lethal disease with a 5-year survival rate of 4%. The only opportunity for improved survival continues to be complete surgical resection for those with localized disease. Although chemotherapeutic options are limited for the few patients with resectable disease, this problem is even more magnified in the majority (85%) of patients with unresectable or metastastic disease. Therefore, there is an urgent need for improved therapeutic options. The recent success of inhibitors of signal transduction for the treatment of other cancers supports the need to identify and validate aberrant signaling pathways important for pancreatic tumor growth. This review focuses on the validation of specific signaling networks and the present status of inhibitors of these pathways as therapeutic approaches for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Jen Jen Yeh
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Division of Surgical Oncology, Chapel Hill, NC 27599, USA.
| | | |
Collapse
|
21
|
Gaspar NJ, Li L, Kapoun AM, Medicherla S, Reddy M, Li G, O'Young G, Quon D, Henson M, Damm DL, Muiru GT, Murphy A, Higgins LS, Chakravarty S, Wong DH. Inhibition of transforming growth factor beta signaling reduces pancreatic adenocarcinoma growth and invasiveness. Mol Pharmacol 2007; 72:152-61. [PMID: 17400764 DOI: 10.1124/mol.106.029025] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor beta (TGFbeta) is a pleiotropic factor that regulates cell proliferation, angiogenesis, metastasis, and immune suppression. Dysregulation of the TGFbeta pathway in tumor cells often leads to resistance to the antiproliferative effects of TGFbeta while supporting other cellular processes that promote tumor invasiveness and growth. In the present study, SD-208, a 2,4-disubstituted pteridine, ATP-competitive inhibitor of the TGFbeta receptor I kinase (TGFbetaRI), was used to inhibit cellular activities and tumor progression of PANC-1, a human pancreatic tumor line. SD-208 blocked TGFbeta-dependent Smad2 phosphorylation and expression of TGFbeta-inducible proteins in cell culture. cDNA microarray analysis and functional gene clustering identified groups of TGFbeta-regulated genes involved in metastasis, angiogenesis, cell proliferation, survival, and apoptosis. These gene responses were inhibited by SD-208. Using a Boyden chamber motility assay, we demonstrated that SD-208 inhibited TGFbeta-stimulated invasion in vitro. An orthotopic xenograft mouse model revealed that SD-208 reduced primary tumor growth and decreased the incidence of metastasis in vivo. Our findings suggest mechanisms through which TGFbeta signaling may promote tumor progression in pancreatic adenocarcinoma. Moreover, they suggest that inhibition of TGFbetaRI with a small-molecule inhibitor may be effective as a therapeutic approach to treat human pancreatic cancer.
Collapse
|
22
|
Aho U, Zhao X, Löhr M, Andersson R. Molecular mechanisms of pancreatic cancer and potential targets of treatment. Scand J Gastroenterol 2007; 42:279-96. [PMID: 17354106 DOI: 10.1080/00365520601106384] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Ursula Aho
- Department of Surgery, Lund University Hospital, University of Lund, Lund, Sweden
| | | | | | | |
Collapse
|
23
|
Truty MJ, Urrutia R. Transforming growth factor-β: What every pancreatic surgeon should know. Surgery 2007; 141:1-6. [PMID: 17188161 DOI: 10.1016/j.surg.2006.07.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2006] [Accepted: 07/10/2006] [Indexed: 12/01/2022]
Affiliation(s)
- Mark J Truty
- Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | |
Collapse
|
24
|
Olive KP, Tuveson DA. The Use of Targeted Mouse Models for Preclinical Testing of Novel Cancer Therapeutics. Clin Cancer Res 2006; 12:5277-87. [PMID: 17000660 DOI: 10.1158/1078-0432.ccr-06-0436] [Citation(s) in RCA: 198] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The use of genetically engineered cancer-prone mice as relevant surrogates for patients during the development of pertinent clinical applications is an unproven expectation that awaits direct demonstration. Despite the generally disappointing findings using tumor xenografts and certain early transgenic cancer models to predict therapeutic efficacy in patients, the dramatic progress of mouse models in recent years engenders optimism that the newest generation of mouse models will provide a higher standard of predictive utility in the process of drug development.
Collapse
Affiliation(s)
- Kenneth P Olive
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
25
|
Gupta P, Su ZZ, Lebedeva IV, Sarkar D, Sauane M, Emdad L, Bachelor MA, Grant S, Curiel DT, Dent P, Fisher PB. mda-7/IL-24: multifunctional cancer-specific apoptosis-inducing cytokine. Pharmacol Ther 2006; 111:596-628. [PMID: 16464504 PMCID: PMC1781515 DOI: 10.1016/j.pharmthera.2005.11.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 02/07/2023]
Abstract
"Differentiation therapy" provides a unique and potentially effective, less toxic treatment paradigm for cancer. Moreover, combining "differentiation therapy" with molecular approaches presents an unparalleled opportunity to identify and clone genes mediating cancer growth control, differentiation, senescence, and programmed cell death (apoptosis). Subtraction hybridization applied to human melanoma cells induced to terminally differentiate by treatment with fibroblast interferon (IFN-beta) plus mezerein (MEZ) permitted cloning of melanoma differentiation associated (mda) genes. Founded on its novel properties, one particular mda gene, mda-7, now classified as a member of the interleukin (IL)-10 gene family (IL-24) because of conserved structure, chromosomal location, and cytokine-like properties has become the focus of attention of multiple laboratories. When administered by transfection or adenovirus-transduction into a spectrum of tumor cell types, melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24) induces apoptosis, whereas no toxicity is apparent in normal cells. mda-7/IL-24 displays potent "bystander antitumor" activity and also has the capacity to enhance radiation lethality, to induce immune-regulatory activities, and to inhibit tumor angiogenesis. Based on these remarkable attributes and effective antitumor therapy in animal models, this cytokine has taken the important step of entering the clinic. In a Phase I clinical trial, intratumoral injections of adenovirus-administered mda-7/IL-24 (Ad.mda-7) was safe, elicited tumor-regulatory and immune-activating processes, and provided clinically significant activity. This review highlights our current understanding of the diverse activities and properties of this novel cytokine, with potential to become a prominent gene therapy for cancer.
Collapse
Affiliation(s)
- Pankaj Gupta
- Department of Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Dang C, Zhang Y, Ma Q, Shimahara Y. Expression of nerve growth factor receptors is correlated with progression and prognosis of human pancreatic cancer. J Gastroenterol Hepatol 2006; 21:850-8. [PMID: 16704535 DOI: 10.1111/j.1440-1746.2006.04074.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM The aim of the present study was to investigate the prognostic value of the two types of nerve growth factor receptors (NGFR), namely high-affinity receptor TrkA and low-affinity receptor p75NGFR, in pancreatic cancer. METHODS The mRNA expression of NGFR for TrkA and p75NGFR was examined in 56 human primary pancreatic cancers using real-time quantitative reverse transcription-polymerase chain reaction. RESULTS Nerve growth factor (NGF) receptors were found in all tumor specimens. It appears that the growth of pancreatic cancer cells stimulated by NGF depended on the expression levels and the ratio of TrkA to p75NGFR. TrkA and p75NGFR were negatively correlated and both were associated with abdominal or back pain and perineural invasion. Regarding this, patients with high TrkA expression levels exhibited more frequent perineural invasion and a higher degree of pain, whereas the results of p75NGFR were opposite. For Cox univariate analyses in the overall survival study, high expression of p75NGFR was associated with longer overall survival, but TrkA exhibited opposite effects and included an effect on perineural invasion and pain. Histoprognostic grading, tumor size and node involvement were not prognostic factors. In Cox multivariate analyses, TrkA and p75NGFR were both prognostic parameters. CONCLUSIONS The present study found that the expression of TrkA in pancreatic cancer is a marker of tumor aggressiveness. Conversely, we also found that elevated p75NGFR expression is associated with a favorable prognosis. We demonstrated that NGF exerts both stimulatory and inhibitory effects on pancreatic cancers, with the overall effect determined by the expression levels and the ratio of TrkA to p75NGFR.
Collapse
Affiliation(s)
- Chengxue Dang
- Department of Surgical Oncology, First Hospital of Xi'an Jiaotong University, Xi'an, China.
| | | | | | | |
Collapse
|
27
|
Comparative analysis of lymphatic metastasis — Associated genes in mouse hepatocellular carcinoma cell lines with different metastatic potential. Chin J Cancer Res 2006. [DOI: 10.1007/s11670-006-0026-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
28
|
Asano T, Yao Y, Shin S, McCubrey J, Abbruzzese JL, Reddy SAG. Insulin receptor substrate is a mediator of phosphoinositide 3-kinase activation in quiescent pancreatic cancer cells. Cancer Res 2005; 65:9164-8. [PMID: 16230374 DOI: 10.1158/0008-5472.can-05-0779] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Phosphoinositide 3-kinase (PI3K) is activated in pancreatic cancer cells and plays a central role in their proliferation, survival, and drug resistance. Although the mechanism is unclear, PI3K activation in these cells could be due to physical interaction between its regulatory subunit (p85) and specific tyrosine kinases or their mediators. Consistent with this possibility, PI3K was precipitated with anti-phosphotyrosine antibodies and Akt phosphorylation was blocked by the tyrosine kinase inhibitors SU6656 and PD158780 in quiescent pancreatic cancer cells. Pull-down assays with a fusion protein (GST-p85NC-SH2), and coimmunoprecipitation studies, indicated that the insulin receptor substrate (IRS), and not the epidermal growth factor and insulin-like growth factor receptors or the Src tyrosine kinase, was physically associated with PI3K in these cells. Our data also indicated that SU6656 and PD158780 inhibited Akt activation in pancreatic cancer cells by interfering with the ability of IRS-1 to recruit PI3K. Furthermore, IRS-1 was phosphorylated on a p85-binding site (Y(612)), and IRS-specific small interfering RNA potently inhibited activation of PI3K and Akt in transfected cells. Taken together, these observations indicate that IRS is a mediator of PI3K activation in quiescent pancreatic cancer cells.
Collapse
Affiliation(s)
- Takayuki Asano
- Department of Gastrointestinal Medical Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, Texas and Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
The prognosis of patients with some kinds of cancers whose patients are often found unresectable upon diagnosis is still dismal. In these fields, development of a new therapeutic modality is needed and gene therapy represents one promising strategy. So far, numerous cancer gene therapy clinical trials based on these principles have been carried out and have shown the safety of such modalities, but have fallen short of the initial expectations to cure cancers. In this review, we would like to make a problem-oriented discussion of current status of cancer gene therapy research by using mainly gastrointestinal cancers as an example. In order to overcome obstacles for full realization of cancer gene therapy, numerous researches have been conducted by many researchers. Various cancer-selective and non-selective genes, as well as lytic viruses themselves have been employed for gene therapy. In the context of gene delivery method, different kinds of viral and non-viral strategies have been utilized. In addition, surrogate assays, such as soluble markers and imaging, have been developed for safer and more informative clinical trials. Many experiments and clinical trials to date have figured out current obstacles for the realization of an effective cancer gene therapy modality. Tireless efforts to overcome such hurdles and continuous infusion of novel concepts into this field should lead to break through technologies and the cure of the patients.
Collapse
Affiliation(s)
- Masato Yamamoto
- BMR2-410, 901 19th Street South, Birmingham, AL 35294-2172, USA
| | | |
Collapse
|
30
|
Li J, Kleeff J, Esposito I, Kayed H, Felix K, Giese T, Büchler MW, Friess H. Expression analysis of PMP22/Gas3 in premalignant and malignant pancreatic lesions. J Histochem Cytochem 2005; 53:885-93. [PMID: 15995147 DOI: 10.1369/jhc.4a6546.2005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
PMP22 is a structural protein of Schwann cells, but it also influences cell proliferation. In the present study, quantitative RT-PCR (QRT-PCR) and immunohistochemistry were used to determine PMP22 mRNA levels and to localize PMP22 in the normal pancreas (n=20), chronic pancreatitis (CP) (n=22), pancreatic ductal adenocarcinoma (PDAC) (n=31), intraductal papillary mucinous neoplasms (IPMN) (n=9), mucinous cystic tumors (MCN) (n=4), and in a panel of PanIN lesions (n=29). PMP22 mRNA levels were significantly higher in CP (3-fold) and PDAC (2.5-fold), compared to normal pancreatic tissues. PMP22 expression was restricted to nerves in the normal pancreas, while in CP and PDAC PMP22 was also expressed in PanIN lesions and in a small percentage of pancreatic cancer cells. PMP22 was weak to absent in the tumor cells of IPMNs and MCNs. PMP22 mRNA was present at different levels in cultured pancreatic cancer cells and up-regulated by transforming growth factor (TGF)-beta1 in 2 of 8 of these cell lines. In conclusion, PMP22 expression is present in both CP and PDAC tissues. Its expression in PanIN lesions and some pancreatic cancer cells in vitro and in vivo suggests a role of PMP22 in the neoplastic transformation process from the normal pancreas to pre-malignant lesions to pancreatic cancer.
Collapse
Affiliation(s)
- Junsheng Li
- Department of General Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The clinical outcome of advanced gastrointestinal (GI) cancers (especially pancreatic and oesophageal cancers) is dismal, despite the advance of conventional therapeutic strategies. Cancer gene therapy is a category of new therapeutics, among which conditionally replicative adenovirus (CRAd) is one promising strategy to overcome existing obstacles of cancer gene therapy. Various CRAds have been developed for GI cancer treatment by taking advantage of the replication biology of adenovirus. Some CRAds have already been tested in clinical trials, but have fallen short of initial expectations. Concerns for clinical applicability include therapeutic potency, replication selectivity and interval end points in clinical trials. In addition, improvement of experimental animal models is needed for a deeper understanding of CRAd biology. Despite these obstacles, CRAds continue to be an exciting area of investigation with great potential for clinical utility. Further virological and oncological research will eventually lead to full realisation of the therapeutic potential of CRAds in the field of GI cancers.
Collapse
Affiliation(s)
- Masato Yamamoto
- Division of Human Gene Therapy, Department of Medicine, and the Gene Therapy Center, University of Alabama at Birmingham, BMR2-408, 901 19th Street South, Birmingham, AL 35294-2172, USA.
| |
Collapse
|
32
|
Song B, Tang JW, Wang B, Cui XN, Hou L, Sun L, Mao LM, Zhou CH, Du Y, Wang LH, Wang HX, Zheng RS, Sun L. Identify lymphatic metastasis-associated genes in mouse hepatocarcinoma cell lines using gene chip. World J Gastroenterol 2005; 11:1463-72. [PMID: 15770722 PMCID: PMC4305688 DOI: 10.3748/wjg.v11.i10.1463] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: In order to obtain lymphogenous metastasis-associated genes, we compared the transcriptional profiles of mouse hepatocarcinoma cell lines Hca-F with highly lymphatic metastasis potential and Hca-P with low lymphatic metastasis potential.
METHODS: Total RNA was isolated from Hca-F and Hca-P cells and synthesized into double-stranded cDNA. In vitro transcription double-stranded cDNA was labeled with biotin (i.e., biotin-labeled cRNA, used as the probe). The cRNA probes hybridized with Affymetrix GeneChip® MOE430A (containing 22690 transcripts, including 14500 known mouse genes and 4371 ESTs) respectively and the signals were scanned by the GeneArray Scanner. The results were then analyzed by bioinformatics.
RESULTS: Out of the 14500 known genes investigated, 110 (0.8%) were up regulated at least 23 fold. Among the total 4371 ESTs, 17 ESTs (0.4%) (data were not presented) were up regulated at least 23 fold. According to the Gene Ontology and TreeView analysis, the 110 genes were further classified into two groups: differential biological process profile and molecular function profile.
CONCLUSION: Using high-throughput gene chip method, a large number of genes and their cellular functions about angiogenesis, cell adhesion, signal transduction, cell motility, transport, microtubule-based process, cytoskeleton organization and biogenesis, cell cycle, transcription, chaperone activity, motor activity, protein kinase activity, receptor binding and protein binding might be involved in the process of lymphatic metastasis and deserve to be used as potential candidates for further investigation. Cyclin D1, Fosl1, Hsp47, EGFR and AR, and Cav-1 are selected as the possible candidate genes of the metastatic phenotype, which need to be validated in later experiments. ESTs (data were not presented) might indicate novel genes associated with lymphatic metastasis. Validating the function of these genes is helpful to identify the key or candidate gene/pathway responsible for lymphatic metastasis, which might be used as the diagnostic markers and the therapeutic targets for lymphatic metastasis.
Collapse
Affiliation(s)
- Bo Song
- Department of Pathology, Dalian Medical University, Dalian 116027, Liaoning Province, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL, Reddy SAG. The PI 3-kinase/Akt signaling pathway is activated due to aberrant Pten expression and targets transcription factors NF-κB and c-Myc in pancreatic cancer cells. Oncogene 2004; 23:8571-80. [PMID: 15467756 DOI: 10.1038/sj.onc.1207902] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The persistent activation of signaling cascades results in dramatic consequences that include loss of cellular growth control and neoplastic transformation. We show here that phosphoinositide 3-kinase (PI 3-kinase) and its mediator Akt were constitutively activated in pancreatic cancer and that this might be due to the aberrant expression of their natural antagonist MMAC/PTEN. Indeed, our results show that MMAC/PTEN expression was either lost or significantly reduced in five of eight cell lines and in twelve of seventeen tumor specimens examined. That the poor expression of MMAC/PTEN in pancreatic cancer cells could be due to promoter methylation was indicated by methylation-specific PCR analysis. Our studies also indicated that PI 3-kinase targeted two important transcription factors in pancreatic cancer cells. The ability of constitutively activated NF-kappaB to induce gene expression and the stabilization of c-MYC protein by decreased phosphorylation of Thr58 were both dependent on PI 3-kinase activity. When pancreatic cancer cells were treated with a peptide antagonist of NF-kappaB nuclear translocation, or stably transfected with a dominant-negative mutant of MYC, their proliferation was markedly inhibited. Taken together, these data indicate that the aberrant expression of MMAC/PTEN contributes to the activation of the PI 3-kinase/Akt pathway and its transcription factor mediators in pancreatic cancer.
Collapse
Affiliation(s)
- Takayuki Asano
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
34
|
Li J, Kleeff J, Kayed H, Felix K, Penzel R, Büchler MW, Korc M, Friess H. Glypican-1 antisense transfection modulates TGF-β-dependent signaling in Colo-357 pancreatic cancer cells. Biochem Biophys Res Commun 2004; 320:1148-55. [PMID: 15249209 DOI: 10.1016/j.bbrc.2004.06.063] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2004] [Indexed: 11/26/2022]
Abstract
The heparan sulfate proteoglycan glypican-1 is essential as a co-receptor for heparin binding growth factors, such as HB-EGF and FGF-2, in pancreatic cancer cells. In the present study, the role of glypican-1 in the regulation of TGF-beta signaling was investigated. Colo-357 pancreatic cancer cells were stably transfected with a full-length glypican-1 antisense construct. Cell growth was determined by MTT and soft agar assays. TGF-beta1 induced p21 expression and Smad2 phosphorylation were analyzed by immunoblotting. PAI-1 promoter activity was determined by luciferase assays. Down-regulation of glypican-1 expression by stable transfection of a full-length glypican-1 antisense construct resulted in decreased anchorage-dependent and -independent cell growth in Colo-357 pancreatic cancer cells and attenuated TGF-beta1 induced cell growth inhibition, Smad2 phosphorylation, and PAI-1 promoter activity. There was, however, no significant difference in TGF-beta1 induced p21 expression and Smad2 nuclear translocation. In conclusion, glypican-1 is required for efficient TGF-beta1 signaling in pancreatic cancer cells.
Collapse
Affiliation(s)
- Junsheng Li
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Affiliation(s)
- Pedro J Ramírez
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
36
|
Raben D, Helfrich B, Bunn PA. Targeted therapies for non–small-cell lung cancer: biology, rationale, and preclinical results from a radiation oncology perspective. Int J Radiat Oncol Biol Phys 2004; 59:27-38. [PMID: 15142632 DOI: 10.1016/j.ijrobp.2004.01.054] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2003] [Revised: 12/30/2003] [Accepted: 01/16/2004] [Indexed: 01/03/2023]
Abstract
The epidermal growth factor receptor (EGFR) is overexpressed in the majority of non-small-cell lung cancers (NSCLCs). This presents an opportune target for new treatment strategies designed to selectively interfere with the cancer cell growth cycle. Recent investigations into the biology of the EGFR and its downstream signaling pathways have reminded us of the complexity of cancer cell communications from the cytoplasm to the nucleus. Multiple pathways are activated with stimulation of the autocrine and paracrine EGFR loop, from the ras-raf-MEK activation of ERK 1/2 to the P13K-Akt pathway, each playing an important role in cancer cell survival, invasion, and angiogenesis. Preclinical studies have demonstrated that molecules targeting the EGFR, either through extracellular blockade or intracellular interference with the EGFR-associated tyrosine kinase, reversibly or irreversibly, inhibit cancer cell growth. Potent antitumor effects have been observed in human tumor xenograft models. Preclinical studies have also demonstrated cooperative effects when anti-EGFR agents are combined with radiation or chemotherapy. Many of these agents have now entered into advanced human clinical trials with modest dose-related toxicity despite chronic administration. Encouraging response rates with single-agent targeted therapy have been reported in heavily pretreated patients with advanced NSCLC. In addition, agents targeting the angiogenic pathway, which plays a key role in the regulation of angiogenesis, may play an important role in enhancing the efficacy of anti-EGFR agents. This article will focus on the biology, rationale, and preclinical studies with targeted anti-EGFR and antiangiogenic therapies for the management of NSCLC.
Collapse
Affiliation(s)
- David Raben
- Department of Radiation Oncology, University of Colorado Health Sciences Center, Denver, USA.
| | | | | |
Collapse
|
37
|
Abstract
Pancreatic adenocarcinoma remains a widespread and difficult disease to treat. Surgical resection offers the possibility of cure in a select few. However, most patients are not eligible, and conventional chemotherapy and radiation remain largely ineffective. Despite this, our understanding of the pathogenesis of the disease has advanced considerably over the past few decades. These findings provide the basis for the development of much needed new therapeutic modalities. Specifically, the application of new recombinant DNA technology and techniques to pancreatic cancer has yielded promising results. This paper reviews our current understanding of the molecular biology of pancreatic adenocarcinoma and its applications to the field of gene therapy.
Collapse
Affiliation(s)
- Pedro J Ramírez
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA.
| | | |
Collapse
|
38
|
Gopalakrishnan VK, Banerjee AG, Vishwanatha JK. Effect of FHIT gene replacement on growth, cell cycle and apoptosis in pancreatic cancer cells. Pancreatology 2004; 3:293-302. [PMID: 12890991 DOI: 10.1159/000071767] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2002] [Accepted: 05/06/2003] [Indexed: 12/11/2022]
Abstract
The human FHIT gene is altered or lost in many cancers and FHIT has been shown to be a tumor suppressor. However, the mechanism of tumor suppression by the FHIT gene remains unclear. FHIT expression is lost in primary pancreatic cancer and human pancreatic cancer cell lines. To gain insight into the function of FHIT gene, we replaced the FHIT gene in a FHIT-null pancreatic cancer cell line, and established stable fhit-expressing clones. Expression of the exogenous fhit was at similar levels as in other cultured cell lines and fhit protein was found predominantly associated with perinuclear area. fhit replacement resulted in reduced cell proliferation in transfected Panc-1 cells. Cell cycle distribution analysis indicated increased accumulation of G(0)/G(1) phase cells in transfected clones indicating a retardation of cell cycle progression. We observed specific up-regulation of cdc2 and cyclin D3 upon fhit replacement. Furthermore, Bcl-2 family members Bad, Bak, and Bcl-xS protein levels were increased in FHIT transfected clones when compared with Panc-1 cells. Multiplex RT-PCR of apoptosis pathway related genes revealed that Bcl-2 is absent and Bcl- xS message increases in FHIT transfected clones. Our data suggested that exogenous expression of FHIT in Panc-1 cells affects genes regulating cell cycle arrest and apoptosis, and these molecular changes may contribute to the tumor suppressor activity of the FHIT gene.
Collapse
Affiliation(s)
- Velliyur K Gopalakrishnan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | |
Collapse
|
39
|
Matsui Y, Asano T, Kenmochi T, Iwakawa M, Imai T, Ochiai T. Effects of carbon-ion beams on human pancreatic cancer cell lines that differ in genetic status. Am J Clin Oncol 2004; 27:24-8. [PMID: 14758129 DOI: 10.1097/01.coc.0000046037.75545.ad] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The relative biologic effectiveness (RBE) of carbon-ion beams at 3 different linear energy transfer (LET) values (13, 50, and 80 keV/microm) accelerated by the Heavy Ion Medical Accelerator in Chiba on human pancreatic cancer cell lines differing in genetic status was determined. The RBE values were calculated as D10, the dose (Gy) required to reduce the surviving fraction to 10%, relative to X-rays. We also investigated apoptosis and the relationship between D10 and the cell cycle checkpoint using morphologic examination and flow cytometry analysis, respectively. The RBE values calculated by the D10 values ranged from 1.16 to 1.77 for the 13-keV/microm beam and from 1.83 to 2.46 for the 80-keV/microm beam. A correlation between the D10 values of each cell line and intensity of G2/M arrest was observed. In contrast, LET values did not clearly correlate with induction of apoptosis. These results suggest that carbon-ion beam therapy is a promising modality. Elucidation of the mechanisms of G2/M arrest and apoptosis may provide clues to enhancing the effects of radiation on pancreatic cancer.
Collapse
Affiliation(s)
- Yoshifumi Matsui
- Frontier Research Center, National Institute of Radiological Sciences, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Tepel J, Kruse ML, March C, Fiedler A, Kapischke M, Ketterer T, Sipos B, Kremer B, Kalthoff H. Terminally modified oligodeoxynucleotides directed against p53 in an orthotopic xenograft model: a novel adjuvant treatment strategy for pancreatic ductal carcinoma. Pancreas 2004; 28:1-12. [PMID: 14707723 DOI: 10.1097/00006676-200401000-00001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Investigation of a terminally modified oligodeoxynucleotide (ODN) directed against p53 mRNA (p53-3' polyethylene glycol-5' tocopherol ODN as a novel drug for pancreatic ductal carcinoma therapy in vitro and in vivo. METHODS The impact of lipophilic modifications at the 5' end of p53-directed ODNs on cellular uptake was analyzed in vitro using proliferation assays, fluorescence-activated cell sorting analysis, and confocal laser scanning microscopy. The in vivo effects of p53-PT-ODN on the growth of orthotopically xenografted human pancreatic ductal carcinoma cells (PancTuI) were studied in SCID beige mice. Distribution was examined in vitro and in vivo using Cy3-labeled ODNs. RESULTS Terminally modified p53-PT-ODN showed excellent cellular uptake without using transfection reagents. Microscopically detectable levels of p53-PT-ODN were reached in vivo within 3 hours after intraperitoneal injection, even in extraperitoneal organs. At this time, Cy3-labeled p53-PT-ODN was found in solid tumor formations. We observed a significant inhibition of tumor growth (50%) in vivo at low doses of p53-PT-ODN, whereas at high doses, 2 of 9 animals had no detectable tumors at necropsy. When p53-PT-ODN was injected on the day of tumor cell inoculation, the growth inhibition of solid tumors was significantly stronger compared with that with delayed treatment. CONCLUSIONS p53-Directed modified ODNs might be of therapeutic value in pancreatic ductal carcinoma, particularly as adjuvant therapy after pancreatic tumor resection.
Collapse
Affiliation(s)
- Jürgen Tepel
- Department of General Surgery, University of Kiel, Kiel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kleeff J, Kothari NH, Friess H, Fan H, Korc M. Adenovirus-mediated transfer of a truncated fibroblast growth factor (FGF) type I receptor blocks FGF-2 signaling in multiple pancreatic cancer cell lines. Pancreas 2004; 28:25-30. [PMID: 14707726 DOI: 10.1097/00006676-200401000-00004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Pancreatic ductal adenocarcinomas (PDACs) overexpress several members of the fibroblast growth factor (FGF) family of ligands and the type I FGF receptor (FGFR-1), and enhanced FGF-2 protein levels correlate with shorter postoperative survival of patients with PDAC. In this study, we investigated the effects of FGF-2 on cell proliferation and mitogen-activated protein kinase (MAPK) activation before and after abrogation of FGFR-1-dependent signaling in 4 pancreatic cancer cell lines (ASPC-1, COLO-357, MIA-PaCa-2, and PANC-1). Signaling was blocked by infecting the cells with an adenoviral vector encoding for a truncated FGFR-1 (AdtrFGFR-1). FGF-2 enhanced the growth of all 4 cell lines and activated MAPK in 3 of these cell lines. Infection with the AdtrFGFR-1 virus resulted in abundant expression of the truncated FGFR-1 at the RNA and protein level, markedly attenuated FGF-2-induced proliferation in all 4 tested cell lines, and decreased FGF-2-dependent MAPK activation in the 3 cell lines in which FGF-2 activated this pathway. These findings suggest that FGFR-1-mediated mitogenesis in multiple pancreatic cancer cells can be efficiently blocked with an adenoviral vector encoding a truncated FGFR-1, raising the possibility that AdtrFGFR-1 may ultimately have a therapeutic role in PDAC.
Collapse
Affiliation(s)
- Jörg Kleeff
- Division of Endocrinology, Department of Medicine, University of Heidelberg, Germany
| | | | | | | | | |
Collapse
|
42
|
Yamamoto S, Nakamori S, Tsujie M, Takahashi Y, Nagano H, Dono K, Umeshita K, Sakon M, Tomita Y, Hoshida Y, Aozasa K, Kohno K, Monden M. Expression of uridine diphosphate N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyl transferase 3 in adenocarcinoma of the pancreas. Pathobiology 2004; 71:12-8. [PMID: 14555840 DOI: 10.1159/000072957] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2003] [Accepted: 03/07/2003] [Indexed: 11/19/2022] Open
Abstract
Uridine diphosphate (UDP) N-acetyl-alpha-D-galactosamine:polypeptide N-acetylgalactosaminyl transferase 3 (GalNAc-T3), one of the enzymes that catalyze the initial glycosylation of mucin type O-linked proteins, was shown to associate with the differentiation of cancer cell lines and the prognosis of some kinds of cancers. In the present study, the association of GalNAc-T3 expression with clinicopathologic features of pancreatic adenocarcinoma and patients' survival was examined. The level of expression of GalNAc-T3 was analyzed immunohistochemically in paraffin-embedded tumor samples from surgically resected specimens of 59 patients with pancreatic ductal adenocarcinoma. The correlations of GalNAc-T3 expression with clinicopathologic features and prognosis were studied. Thirty-five tumors showed high- intensity GalNAc-T3 staining, whereas 24 showed low-intensity staining. A close association was observed between GalNAc-T3 staining intensity and histologic differentiation and the stage of the tumors. The low-intensity group showed a high rate of the poorly differentiated subtype (p < 0.001) and T4 of the pTNM staging system (p < 0.05). These findings indicate that the expression of GalNAc-T3 is associated with the differentiation and aggressiveness of ductal adenocarcinoma of the pancreas.
Collapse
Affiliation(s)
- Shinji Yamamoto
- Department of Surgery and Clinical Oncology, Osaka University Graduate School of Medicine, Fukuoka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Pancreatic cancer is an almost universally fatal disease, with a five-year survival rate of 5%. Research into both sporadic and inherited forms of pancreatic cancer has yielded tremendous advances in the understanding of this disease at the molecular level. Elucidating genetic alterations in pancreatic cancer has identified various abnormalities ranging from gross chromosomal abnormalities to point mutations, many of which influence the development and progression of pancreatic cancer. Identifying precursor lesions within pancreatic ducts has led to the formulation of a progression model of pancreatic cancer and subsequent identification of early- and late-stage changes leading to invasive cancer. Ultimately, understanding the genetic events underlying the development of pancreatic cancer may serve as a useful adjunct in the screening and treatment of patients suffering from, or at risk for, pancreatic cancer.
Collapse
Affiliation(s)
- Donna E Hansel
- Department of Pathology, The Johns Hopkins Medical Institutions, Baltimore, Maryland 21231, USA.
| | | | | |
Collapse
|
44
|
Huang ZQ, Buchsbaum DJ, Raisch KP, Bonner JA, Bland KI, Vickers SM. Differential responses by pancreatic carcinoma cell lines to prolonged exposure to Erbitux (IMC-C225) anti-EGFR antibody. J Surg Res 2003; 111:274-83. [PMID: 12850474 DOI: 10.1016/s0022-4804(03)00076-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Pancreatic cancer remains a devastating disease, with 95% of all patients diagnosed with the disease dying within 2 years. The combined therapy using Erbitux, gemcitabine, and radiation caused complete tumor regression using a nude mouse model inoculated with pancreatic MiaPaCa-2 cells but only a delay in tumor growth with BxPC-3. We investigated the effect of prolonged Erbitux treatment to the sensitivity to gemcitabine and/or radiation and the epidermal growth factor receptor (EGFR) signal transduction pathway. METHODS MiaPaCa-2 and BxPC-3 cells were cultured with or without Erbitux for 6 weeks. Cells were then treated with gemcitabine and/or radiation, harvested 48 h after treatment, and counted. Differences in EGFR expression after exposure to Erbitux were analyzed by FACS. Internalization rates of EGFR induced by Erbitux on these cell lines were determined using 125I-EGF binding assay after removal of Erbitux by acidic wash. Cell lysates were harvested after cells were stimulated with EGF, FGF, or IGF-1 respectively, and EGFR was immunoprecipitated using Erbitux. Samples were separated using SDS-PAGE and transferred to PVDF membrane. The membranes were probed with antibody against human growth factor receptor binding protein (Grb2) to detect the association of this Ras-MAPK upstream adaptor protein to EGFR. Cell lysates were also separated with SDS-PAGE and probed with rabbit anti-human PARP after samples were transferred to PVDF membrane. Expression of BAX and Bcl-(XL) were probed in the cells treated with or without Erbitux. RESULTS Proliferation assays indicated that prolonged exposure to Erbitux increased the sensitivities of MiaPaCa-2 to gemcitabine and radiation therapy (41 +/- 16% vs 52 +/- 9% for gemcitation, 28 +/- 9 vs 39 +/- 9% for combination; P = 0.015) but not for BxPC-3. FACS analysis showed that the expressed EGFR level decreased by about 42% on MiaPaCa 2 whereas no loss was seen on BxPC-3. Expression of BAX was upregulated on MiaPaCa-2. Poly (ADP-ribose) polymerase cleavage indicated the killing was mediated by apoptosis. Immunoblots showed that Grb2 was co-immunoprecipitated with EGFR after EGF stimulation. Incubation with Erbitux blocked Grb2 binding in MiaPaCa-2 but not BxPC 3. FGF transactivated EGFR down stream Ras-MAPK in the presence or absence of Erbitux. Internalization of EGFR induced by Erbitux did not differ between MiaPaCa-2 and BxPC-3. CONCLUSIONS 1) Association of Grb2 to EGFR in BxPC-3 induced by EGF in the presence of Erbitux indicates an alternate pathway of Ras-MAPK activation, which may be related with the tumor resistance to treatment; 2) transactivation of EGFR downstream Ras-MAPK pathway by FGF contributes the resistance to treatment; and 3) downregulation of EGFR may increase the response to therapy.
Collapse
Affiliation(s)
- Zhi-qiang Huang
- Division of General Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama 35294-0016, USA
| | | | | | | | | | | |
Collapse
|
45
|
Shi M, Wang FS, Wu ZZ. Synergetic anticancer effect of combined quercetin and recombinant adenoviral vector expressing human wild-type p53, GM-CSF and B7-1 genes on hepatocellular carcinoma cells in vitro. World J Gastroenterol 2003; 9:73-8. [PMID: 12508355 PMCID: PMC4728253 DOI: 10.3748/wjg.v9.i1.73] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: This study investigated the anti-cancer effect of combined quercetin and a recombinant adenovirus vector expressing the human p53, GM-CSF and B7-1 genes (designated BB-102) on human hepatocellular carcinoma (HCC) cell lines in vitro.
METHODS: The sensitivity of HCC cells to anticancer agents was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. The viability of cells infected with BB-102 was determined by trypan blue exclusion. The expression levels of human wild-type p53, GM-CSF and B7-1 genes were determined by Western blot, enzyme-linked immunosorbent assay (ELISA) and flow cytometric analysis, respectively. The apoptosis of BB-102-infected or quercetin-treated HCC cells was detected by terminal deoxynucleotidyl transferase (TdT) assay or DNA ladder electrophoresis.
RESULTS: Quercetin was found to suppress proliferation of human HCC cell lines BEL-7402, HuH-7 and HLE, with peak suppression at 50 μmol/L quercetin. BB-102 infection was also found to significantly suppress proliferation of HCC cell lines. The apoptosis of BB-102-infected HCC cells was greater in HLE and HuH-7 cells than in BEL-7402 cells. Quercetin did not affect the expression of the three exogenous genes in BB-102-infected HCC cells (P > 0.05), but it was found to further decrease proliferation and promote apoptosis of BB-102-infected HCC cells.
CONCLUSION: BB-102 and quercetin synergetically suppress HCC cell proliferation and induce HCC cell apoptosis, suggesting a possible use as a combined anti-cancer agent.
Collapse
Affiliation(s)
- Ming Shi
- Division of Biological Engineering, Institute of Infectious Disease, the 302 Hospital of PLA, 26 Fengtai Lu, Beijing 100039, China
| | | | | |
Collapse
|
46
|
Guillermet J, Saint-Laurent N, Rochaix P, Cuvillier O, Levade T, Schally AV, Pradayrol L, Buscail L, Susini C, Bousquet C. Somatostatin receptor subtype 2 sensitizes human pancreatic cancer cells to death ligand-induced apoptosis. Proc Natl Acad Sci U S A 2003; 100:155-60. [PMID: 12490654 PMCID: PMC140912 DOI: 10.1073/pnas.0136771100] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Somatostatin receptor subtype 2 (sst2) gene expression is lost in 90% of human pancreatic adenocarcinomas. We previously demonstrated that stable sst2 transfection of human pancreatic BxPC-3 cells, which do not endogenously express sst2, inhibits cell proliferation, tumorigenicity, and metastasis. These sst2 effects occur as a consequence of an autocrine sst2-dependent loop, whereby sst2 induces expression of its own ligand, somatostatin. Here we investigated whether sst2 induces apoptosis in sst2-transfected BxPC-3 cells. Expression of sst2 induced a 4.4- +/- 0.05-fold stimulation of apoptosis in BxPC-3 through the activation of tyrosine phosphatase SHP-1. sst2 also sensitized these cells to apoptosis induced by tumor necrosis factor alpha (TNFalpha), enhancing it 4.1- +/- 1.5-fold. Apoptosis in BxPC-3 cells mediated by TNF-related apoptosis-inducing ligand (TRAIL) and CD95L was likewise increased 2.3- +/- 0.5-fold and 7.4- +/- 2.5-fold, respectively. sst2-dependent activation and cell sensitization to death ligand-induced apoptosis involved activation of the executioner caspases, key factors in both death ligand- or mitochondria-mediated apoptosis. sst2 affected both pathways: first, by up-regulating expression of TRAIL and TNFalpha receptors, DR4 and TNFRI, respectively, and sensitizing the cells to death ligand-induced initiator capase-8 activation, and, second, by down-regulating expression of the antiapoptotic mitochondrial Bcl-2 protein. These results are of interest for the clinical management of chemoresistant pancreatic adenocarcinoma by using a combined gene therapy based on the cotransfer of genes for both the sst2 and a nontoxic death ligand.
Collapse
Affiliation(s)
- Julie Guillermet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U531, Institut Federatif de Recherche (IFR) 31, Centre Hospitalier Universitaire (CHU) Rangueil, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Nomura H, Nishimori H, Yasoshima T, Hata F, Tanaka H, Nakajima F, Honma T, Araya J, Kamiguchi K, Isomura H, Sato N, Denno R, Hirata K. A new liver metastatic and peritoneal dissemination model established from the same human pancreatic cancer cell line: analysis using cDNA macroarray. Clin Exp Metastasis 2002; 19:391-9. [PMID: 12198767 DOI: 10.1023/a:1016370532618] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
To elucidate the mechanisms of metastasis, we established two sublines HPC-1H5 with a highly liver metastatic cell line and HPC-1P5a with a highly peritoneal disseminating cell line, which were sequentially selected from the parental pancreatic cancer cell line HPC-1. Using these three cell lines, we investigated several biological properties and mRNA levels of differentially-expressed genes involved in cancer metastasis by cDNA macroarray. Microscopic findings for the three cell lines were the same. The tumorigenicity, in vitro growth ability, motile activity, adhesive activity and the production of IL-8 of metastatic sublines were higher than those of parental HPC-1 cells. Particularly, HPC-1H5 cells showed clearly higher levels of IL-8 expression and tumors of HPC-1H5 cells grew faster and bigger than those of HPC-1P5a cells. In cDNA macroarray analysis of HPC-1H5 cells, 22 genes were up-regulated and 44 genes were down-regulated compared with parental HPC-1 cells. In HPC-1P5a cells, 9 genes were up-regulated and 28 genes were down-regulated compared with parental HPC-1 cells. This study provides a demonstration of global gene expression analysis of pancreatic cancer cells with liver metastasis and peritoneal dissemination. Furthermore, our results provide a new insight into the study of liver metastasis and peritoneal dissemination of human pancreatic cancer.
Collapse
Affiliation(s)
- Hiroki Nomura
- First Department of Surgery, Sapporo Medical University School of Medicine, South 1, West 16, Chuo-ku, Sapporo, 060-8543, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Raben D, Bianco C, Helfrich B, Weng E, Ciardiello F, Harari P. Interference with EGFR signaling: paradigm for improving radiation response in cancer treatment. Expert Rev Anticancer Ther 2002; 2:461-71. [PMID: 12647989 DOI: 10.1586/14737140.2.4.461] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The introduction of biologically active agents that interfere with the epidermal growth factor receptor (EGFR) provides a promising opportunity to improve cancer treatment outcomes. Several EGFR-selective agents, such as humanized monoclonal antibodies and small molecule, orally available tyrosine kinase inhibitors have shown antitumor activity in early clinical trials in advanced cancer patients. Preclinical studies have demonstrated enhanced radiation- and chemotherapy-induced tumor cytotoxicity when EGFR antagonists are implemented. More broadly, recent clinical trials have confirmed improved survival with combinations of HER-2 (a member of the ErbB family of receptors) targeted antibodies and chemotherapy in patients with advanced breast cancer. A landmark trial combining C225 antiEGFR antibody with radiation therapy for patients with locally advanced head and neck cancer has just completed accrual. Thus, emerging rapidly are cancer treatment strategies based on an improved understanding of the specific cellular and molecular abnormalities of individual tumors.
Collapse
Affiliation(s)
- David Raben
- University of Colorado Health Sciences Center, Anschutz Cancer Pavilion, Department of Radiation Oncology, Aurora 80010-0510, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
Gene transfer technology has the potential to revolutionize cancer treatment. Developments in molecular biology, genetics, genomics, stem cell technology, virology, bioengineering, and immunology are accelerating the pace of innovation and movement from the laboratory bench to the clinical arena. Pancreatic adenocarcinoma, with its particularly poor prognosis and lack of effective traditional therapy for most patients, is an area where gene transfer and immunotherapy have a maximal opportunity to demonstrate efficacy. In this review, we have discussed current preclinical and clinical investigation of gene transfer technology for pancreatic cancer. We have emphasized that the many strategies under investigation for cancer gene therapy can be classified into two major categories. The first category of therapies rely on the transduction of cells other than tumor cells, or the limited transduction of tumor tissue. These therapies, which do not require efficient gene transfer, generally lead to systemic biological effects (e.g., systemic antitumor immunity, inhibition of tumor angiogenesis, etc) and therefore the effects of limited gene transfer are biologically "amplified." The second category of gene transfer strategies requires the delivery of therapeutic genetic material to all or most tumor cells. While these elegant approaches are based on state-of-the-art advances in our understanding of the molecular biology of cancer, they suffer from the current inadequacies of gene transfer technology. At least in the short term, it is very likely that success in pancreatic cancer gene therapy will involve therapies that require only the limited transduction of cells. The time-worn surgical maxim, "Do what's easy first," certainly applies here.
Collapse
Affiliation(s)
- Jennifer F Tseng
- Division of Molecular Medicine, Children's Hospital, Department of Genetics, Harvard Medical School, Enders 861, 320 Longwood Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
50
|
Cunningham CC. New Modalities in Oncology: Antisense Oligonucleotides. Proc (Bayl Univ Med Cent) 2002; 15:125-8. [PMID: 16333420 PMCID: PMC1276497 DOI: 10.1080/08998280.2002.11927826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Affiliation(s)
- C Casey Cunningham
- Mary Crowley Medical Research Center and the Department of Oncology, Baylor University Medical Center, Dallas, Texas.
| |
Collapse
|