1
|
Singh S, Kaushik AC, Gupta H, Jhinjharia D, Sahi S. Identification of Prognostic Markers and Potential Therapeutic Targets using Gene Expression Profiling and Simulation Studies in Pancreatic Cancer. Curr Comput Aided Drug Des 2024; 20:955-973. [PMID: 37711100 DOI: 10.2174/1573409920666230914100826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/07/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has a 5-year relative survival rate of less than 10% making it one of the most fatal cancers. A lack of early measures of prognosis, challenges in molecular targeted therapy, ineffective adjuvant chemotherapy, and strong resistance to chemotherapy cumulatively make pancreatic cancer challenging to manage. OBJECTIVE The present study aims to enhance understanding of the disease mechanism and its progression by identifying prognostic biomarkers, potential drug targets, and candidate drugs that can be used for therapy in pancreatic cancer. METHODS Gene expression profiles from the GEO database were analyzed to identify reliable prognostic markers and potential drug targets. The disease's molecular mechanism and biological pathways were studied by investigating gene ontologies, KEGG pathways, and survival analysis to understand the strong prognostic power of key DEGs. FDA-approved anti-cancer drugs were screened through cell line databases, and docking studies were performed to identify drugs with high affinity for ARNTL2 and PIK3C2A. Molecular dynamic simulations of drug targets ARNTL2 and PIK3C2A in their native state and complex with nilotinib were carried out for 100 ns to validate their therapeutic potential in PDAC. RESULTS Differentially expressed genes that are crucial regulators, including SUN1, PSMG3, PIK3C2A, SCRN1, and TRIAP1, were identified. Nilotinib as a candidate drug was screened using sensitivity analysis on CCLE and GDSC pancreatic cancer cell lines. Molecular dynamics simulations revealed the underlying mechanism of the binding of nilotinib with ARNTL2 and PIK3C2A and the dynamic perturbations. It validated nilotinib as a promising drug for pancreatic cancer. CONCLUSION This study accounts for prognostic markers, drug targets, and repurposed anti-cancer drugs to highlight their usefulness for translational research on developing novel therapies. Our results revealed potential and prospective clinical applications in drug targets ARNTL2, EGFR, and PI3KC2A for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Samvedna Singh
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | | | - Himanshi Gupta
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Divya Jhinjharia
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| | - Shakti Sahi
- School of Biotechnology, Gautam Buddha University, Greater Noida, India
| |
Collapse
|
2
|
Yoshii Y, Matsumoto H, Igarashi C, Tachibana T, Hihara F, Shinada M, Waki A, Yoshida S, Naito K, Ito K, Higashi T, Kurihara H, Ueno M. Process to Remove the Size Variants Contained in the Antibody-Chelator Complex PCTA-NCAB001 for Radiolabeling with Copper-64. Pharmaceuticals (Basel) 2023; 16:1341. [PMID: 37895812 PMCID: PMC10610008 DOI: 10.3390/ph16101341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
Understanding the physicochemical properties of antibody-drug conjugates is critical to assess their quality at manufacturing and monitor them during subsequent storage. For radiometal-antibody complexes, it is important to control the properties of the antibody-chelator conjugate to maintain the quality of the final product. We have been developing 64Cu-labeled anti-epidermal growth factor receptor antibody NCAB001 (64Cu-NCAB001) for the early diagnosis and therapy of pancreatic cancer with positron-emission tomography. Here, we characterized the larger size variants contained in the antibody-chelator conjugate PCTA-NCAB001 by multi-angle light scattering coupled with size-exclusion chromatography. Secondly, we developed a chromatographic method to remove these size variants. Lastly, we demonstrated the stability of PCTA-NCAB001 after the removal of size variants. Dimer and oligomers were identified in PCTA-NCAB001. These larger size variants, together with some smaller size variants, could be removed by hydrophobic interaction chromatography. The PCTA-NCAB001 product, after the removal of these size variants, could be stored at 4 °C for six months. The methods developed here can be applied to assure the quality of PCTA-NCAB001 and other antibody-drug conjugates to facilitate the development of antibody-radiometal conjugates for positron-emission tomography and radioimmunotherapy of malignant cancers.
Collapse
Affiliation(s)
- Yukie Yoshii
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Hiroki Matsumoto
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Chika Igarashi
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Tomoko Tachibana
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Biology, Graduate School of Science, Toho University, Chiba 274-8510, Japan
| | - Fukiko Hihara
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Mitsuhiro Shinada
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
- Department of Chemistry, Graduate School of Science, Toho University, Chiba 274-8510, Japan
| | - Atsuo Waki
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Sei Yoshida
- Department of Research, NanoCarrier Co., Ltd., Tokyo 104-0031, Japan; (S.Y.); (K.N.)
| | - Kenichiro Naito
- Department of Research, NanoCarrier Co., Ltd., Tokyo 104-0031, Japan; (S.Y.); (K.N.)
| | - Kimiteru Ito
- Department of Diagnostic Radiology, National Cancer Center Hospital, Tokyo 104-0045, Japan;
| | - Tatsuya Higashi
- Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan; (H.M.); (C.I.); (T.T.); (F.H.); (M.S.); (A.W.); (T.H.)
| | - Hiroaki Kurihara
- Department of Diagnostic Radiology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama 241-8515, Japan;
| |
Collapse
|
3
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
4
|
Xie D, Xu P, Yuan C. miR-21 Regulates the Growth of Gastric Cancer Cells Through Targeting Phosphatase and Tensin Homolog (PTEN). J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Gastric cancer (GC) is the leading cause of death worldwide and the prognosis remains poor. Proliferation and apoptosis of cancer cells are regulated by microRNAs (miRNAs). We herein intended to explore the interaction of miR-21 and PTEN in GC. miR-21 inhibitor or negative control was
transfected into GC cells MGC-803 followed by analysis of miR-21 and PTEN level by RT-qPCR, PTEN protein level by western blot and cell growth by MTT and Hoechest-33342 staining. Treatment with miR-21 inhibitor reduced miR-21 expression and increased PTEN protein expression. miR-21 was negatively
associated with PTEN level. Moreover, downregulation of miR-21 decreased cell proliferation and promoted apoptosis. In conclusion, miR-21 stimulates the malignant phenotypes of GC cells by negatively regulating PTEN expression, providing novel insight into the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Dongfang Xie
- Department of Abdominal Tumor Surgery, Qingdao Central Hospital, Qingdao, Shandong, 266042, China
| | - Peng Xu
- Department of Traumatology and Orthopedics, Bone Disease and Bone Tumor, Emergency Surgery, Qingdao Central Hospital, Qingdao, Shandong, 266042, China
| | - Chen Yuan
- Qingdao Central Hospital Health Management Center, Qingdao, Shandong, 266042, China
| |
Collapse
|
5
|
Yao H, Song W, Cao R, Ye C, Zhang L, Chen H, Wang J, Shi Y, Li R, Li Y, Liu X, Zhou X, Shao R, Li L. An EGFR/HER2-targeted conjugate sensitizes gemcitabine-sensitive and resistant pancreatic cancer through different SMAD4-mediated mechanisms. Nat Commun 2022; 13:5506. [PMID: 36127339 PMCID: PMC9489697 DOI: 10.1038/s41467-022-33037-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
Chemoresistance limits its clinical implementation for pancreatic ductal adenocarcinoma (PDAC). We previously generated an EGFR/HER2 targeted conjugate, dual-targeting ligand-based lidamycin (DTLL), which shows a highly potent antitumor effect. To overcome chemoresistance in PDAC, we aim to study DTLL efficacy when combined with gemcitabine and explore its mechanisms of action. DTLL in combination with gemcitabine show a superior inhibitory effect on the growth of gemcitabine-resistant/sensitive tumors. DTLL sensitizes gemcitabine efficacy via distinct action mechanisms mediated by mothers against decapentaplegic homolog 4 (SMAD4). It not only prevents neoplastic proliferation via ATK/mTOR blockade and NF-κB impaired function in SMAD4-sufficient PDACs, but also restores SMAD4 bioactivity to trigger downstream NF-κB-regulated signaling in SMAD4-deficient tumors and to overcome chemoresistance. DTLL seems to act as a SMAD4 module that normalizes its function in PDAC, having a synergistic effect in combination with gemcitabine. Our findings provide insight into a rational SMAD4-directed precision therapy in PDAC. Chemoresistance is a main limitation for the treatment of pancreatic ductal adenocarcinoma (PDAC). Here, the authors show that an antibody drug conjugate-like compound targeting both EGFR and HER2 overcomes gemcitabine resistance in PDAC preclinical models by mechanisms involving the tumour suppressor SMAD4.
Collapse
Affiliation(s)
- Hongjuan Yao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Wenping Song
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China.,Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, No.127 Dongming Road, Zhengzhou, 450008, China
| | - Rui Cao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China.,Academy of Life Science, North China University of Science and Technology, Tangshan, 063210, P. R. China
| | - Cheng Ye
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China.,Tianjin Municipal Health Commission, Tianjin, 300000, P. R. China
| | - Li Zhang
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Hebing Chen
- Beijing Institute of Radiation Medicine, Beijing, 100850, P.R. China
| | - Junting Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P.R. China
| | - Yuchen Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, No.5 Haiyuncang, Beijing, 100700, China
| | - Rui Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Yi Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Xiujun Liu
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Xiaofei Zhou
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China
| | - Rongguang Shao
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China.
| | - Liang Li
- Key Laboratory of Antibiotic Bioengineering of National Health and Family Planning Commission (NHFPC), Institute of Medicinal Biotechnology (IMB), Chinese Academy of Medical Sciences and Peking Union Medical College (CAMS & PUMC), NO.1 TiantanXili, Beijing, 100050, P.R. China.
| |
Collapse
|
6
|
Preclinical Safety Evaluation of Intraperitoneally Administered Cu-Conjugated Anti-EGFR Antibody NCAB001 for the Early Diagnosis of Pancreatic Cancer Using PET. Pharmaceutics 2022; 14:pharmaceutics14091928. [PMID: 36145676 PMCID: PMC9504034 DOI: 10.3390/pharmaceutics14091928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Detecting tumor lesions <1 cm in size using current imaging methods remains a clinical challenge, especially in pancreatic cancer. Previously, we developed a method to identify pancreatic tumor lesions ≥3 mm using positron emission tomography (PET) with an intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor (EGFR) antibody (64Cu-NCAB001 ipPET). Here, we conducted an extended single-dose toxicity study of 64Cu-NCAB001 ipPET in mice based on approach 1 of the current ICH M3 [R2] guideline, as our new drug formulation contains 45 μg of the antibody. We used NCAB001 labeled with stable copper isotope instead of 64Cu. The total content of size variants was approximately 6.0% throughout the study. The relative binding potency of Cu-NCAB001 to recombinant human EGFR was comparable to that of cetuximab. The general and neurological toxicities of Cu-NCAB001 ipPET at 62.5 or 625 μg/kg were assessed in mice. The no-observed-adverse-effect level of Cu-NCAB001 was 625 μg/kg, a dose approximately 1000-fold higher at the μg/kg level than the dose of 64Cu-NCAB001 in our formulation (45 µg). The size variants did not affect the safety of the formulation. Therefore, clinical studies on the efficacy of 64Cu-NCAB001 ipPET for early detection of pancreatic cancer using PET imaging can be safely conducted.
Collapse
|
7
|
Characterization and Stabilization of a New 64Cu-Labeled Anti-EGFR Antibody NCAB001 for the Early Detection of Pancreatic Cancer with Positron Emission Tomography. Pharmaceutics 2021; 14:pharmaceutics14010067. [PMID: 35056963 PMCID: PMC8779674 DOI: 10.3390/pharmaceutics14010067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/24/2021] [Accepted: 12/25/2021] [Indexed: 12/20/2022] Open
Abstract
Early diagnosis of pancreatic cancer using current imaging modalities remains challenging. We have developed a new approach to identify tumor lesions ≥ 3 mm in the pancreas by positron emission tomography (PET) with a new intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor (EGFR) antibody (encoded as NCAB001), called 64Cu-NCAB001 ipPET. Generally, in clinical research, a radiometal-antibody complex must be prepared immediately before use at the imaging site. To make 64Cu-NCAB001 ipPET available to daily clinical practices in a sustainable way, the NCAB001-chelator conjugate and 64Cu-NCAB001 must be characterized and stabilized. NCAB001 was manufactured under cGMP conditions. NCAB001 was conjugated with a bifunctional chelator (p-SCN-Bn-PCTA), and the antibody-chelator conjugate (PCTA-NCAB001) was characterized by LC/MS and ELISA. Thereafter, to effectively manufacture 64Cu-NCAB001, we developed a new formulation to stabilize PCTA-NCAB001 and 64Cu-NCAB001. An average of three PCTA chelators were conjugated per molecule of NCAB001. The relative binding potency of PCTA-NCAB001 was comparable to cetuximab. The formulation consisting of acetate buffer, glycine, and polysorbate-80 stabilized PCTA-NCAB001 for a year-long storage. Additionally, this formulation enabled the stabilization of 64Cu-NCAB001 for up to 24 h after radiolabeling with a sufficient radioactivity concentration for clinical use. These results may accelerate the future use of 64Cu-NCAB001 ipPET in clinical settings for the early diagnosis and treatment of pancreatic cancer.
Collapse
|
8
|
Igarashi C, Yoshii Y, Tashima H, Iwao Y, Sakurai K, Hihara F, Tachibana T, Yoshida E, Wakizaka H, Akamatsu G, Yamaya T, Yoshimoto M, Matsumoto H, Zhang MR, Nagatsu K, Sugyo A, Tsuji AB, Higashi T. Usefulness of PET-guided surgery with 64Cu-labeled cetuximab for resection of intrapancreatic residual tumors in a xenograft mouse model of resectable pancreatic cancer. Nucl Med Commun 2021; 42:1112-1121. [PMID: 34100794 DOI: 10.1097/mnm.0000000000001442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In pancreatic cancer surgery, accurate identification and resection of intrapancreatic residual tumors are quite difficult. We have developed a novel open-typed PET system (called 'OpenPET'), which enables high-resolution PET-guided surgery in real time, and demonstrated that OpenPET-guided surgery with intraperitoneally administered 64Cu-labeled anti-epidermal growth factor receptor antibody cetuximab is useful to detect and resect primary pancreatic cancer. Here, we investigated applicability of OpenPET-guided surgery for unexpected residual intrapancreatic tumors and examined its survival benefit over conventional surgery. METHODS A mouse model with large (>1 cm) resectable pancreatic cancer of xPA-1-DC cells expressing red fluorescent protein was used. OpenPET-guided surgery was conducted 24 h after intraperitoneal administration of 64Cu-labeled cetuximab (7.4 MBq/mouse). For comparison, similar surgical procedures were conducted, and conventional tumor resection was attempted using only the naked eye (control). Survival rate after OpenPET-guided surgery was compared to that after control operations. RESULTS Intraoperative OpenPET guidance enabled detection and resection of small residual tumors. Ten residual tumor specimens (3-10 mm in diameter) were intraoperatively isolated with OpenPET guidance (n = 7 mice). All isolated specimens showed tumor RFP signals. No resection of tumor tissue was performed in control group because the tumor could not be clearly detected with the naked eye alone. Mice after OpenPET-guided surgery showed significantly longer survival rates than those in control group. CONCLUSIONS OpenPET-guided surgery with 64Cu-labeled-cetuximab enabled intraoperative identification and resection of intrapancreatic small residual tumors. This technology could be useful to prevent tumor residuals during surgery and improve pancreatic cancer survival.
Collapse
Affiliation(s)
| | - Yukie Yoshii
- Department of Molecular Imaging and Theranostics
| | - Hideaki Tashima
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Yuma Iwao
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | | | | | | | - Eiji Yoshida
- Department of Molecular Imaging and Theranostics
| | - Hidekatsu Wakizaka
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Go Akamatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Taiga Yamaya
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Mitsuyoshi Yoshimoto
- Division of Functional Imaging, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | | | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Kotaro Nagatsu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Science, National Institutes for Quantum and Radiological Science and Technology, Chiba
| | - Aya Sugyo
- Department of Molecular Imaging and Theranostics
| | | | | |
Collapse
|
9
|
Evaluation of 64Cu-Labeled New Anti-EGFR Antibody NCAB001 with Intraperitoneal Injection for Early PET Diagnosis of Pancreatic Cancer in Orthotopic Tumor-Xenografted Mice and Nonhuman Primates. Pharmaceuticals (Basel) 2021; 14:ph14100950. [PMID: 34681174 PMCID: PMC8540406 DOI: 10.3390/ph14100950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/14/2023] Open
Abstract
Objectives: To improve the prognosis of pancreatic cancer, new imaging methods to identify tumor lesions at a size of <1 cm are urgently needed. To approach this clinical issue, we developed a new method to detect small tumor lesions in the pancreas (≥3 mm) by positron emission tomography (PET) using an intraperitoneally (ip)-administered 64Cu-labeled new anti-epidermal growth factor receptor (EGFR) antibody (encoded as NCAB001), called 64Cu-NCAB001 ipPET. Methods: NCAB001 was manufactured under cGMP conditions and labeled with 64Cu. The radiochemical and biological properties of 64Cu-NCAB001 were evaluated. Tumor uptake of an ip-administered 64Cu-NCAB001 in mice with orthotopic pancreatic tumor xPA1-DC xenografts was also evaluated. Pharmacokinetics and radiation dosimetry were examined using PET images acquired after the ip administration of 64Cu-NCAB001 into cynomolgus monkeys with pharmacologic safety monitoring. Results: Radio-chromatography, cell-binding assays, and biodistribution of 64Cu-NCAB001 in mice were identical to those of our previous data with clinically available cetuximab. Small tumor lesions in the pancreas (≥3 mm) of mice could be identified by 64Cu-NCAB001 ipPET. The ip administration of 64Cu-NCAB001 into monkeys was safely conducted using ultrasound imaging. PET images in monkeys showed that ip-administered 64Cu-NCAB001 was distributed throughout the intraperitoneal cavity for up to 6 h and cleared thereafter. Most of the radioactivity was distributed in the liver and the large intestine. The radioactivity around the pancreas became negligible 24 h after administration. The estimated human effective dose was 0.0174 mSv/MBq. Conclusion: Our data support the initiation of clinical trials of 64Cu-NCAB001 ipPET to transfer this promising tool for the early diagnosis of pancreatic cancers.
Collapse
|
10
|
Garcia-Sampedro A, Gaggia G, Ney A, Mahamed I, Acedo P. The State-of-the-Art of Phase II/III Clinical Trials for Targeted Pancreatic Cancer Therapies. J Clin Med 2021; 10:566. [PMID: 33546207 PMCID: PMC7913382 DOI: 10.3390/jcm10040566] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is a devastating disease with very poor prognosis. Currently, surgery followed by adjuvant chemotherapy represents the only curative option which, unfortunately, is only available for a small group of patients. The majority of pancreatic cancer cases are diagnosed at advanced or metastatic stage when surgical resection is not possible and treatment options are limited. Thus, novel and more effective therapeutic strategies are urgently needed. Molecular profiling together with targeted therapies against key hallmarks of pancreatic cancer appear as a promising approach that could overcome the limitations of conventional chemo- and radio-therapy. In this review, we focus on the latest personalised and multimodal targeted therapies currently undergoing phase II or III clinical trials. We discuss the most promising findings of agents targeting surface receptors, angiogenesis, DNA damage and cell cycle arrest, key signalling pathways, immunotherapies, and the tumour microenvironment.
Collapse
Affiliation(s)
| | | | | | | | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, London NW3 2QG, UK; (A.G.-S.); (G.G.); (A.N.); (I.M.)
| |
Collapse
|
11
|
Jentzsch V, Davis JAA, Djamgoz MBA. Pancreatic Cancer (PDAC): Introduction of Evidence-Based Complementary Measures into Integrative Clinical Management. Cancers (Basel) 2020; 12:E3096. [PMID: 33114159 PMCID: PMC7690843 DOI: 10.3390/cancers12113096] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
The most common form of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC), which comprises some 85% of all cases. Currently, this is the fourth highest cause of cancer mortality worldwide and its incidence is rising steeply. Commonly applied clinical therapies offer limited chance of a lasting cure and the five-year survival rate is one of the lowest of the commonly occurring cancers. This review cultivates the hypothesis that the best management of PDAC would be possible by integrating 'western' clinical medicine with evidence-based complementary measures. Protecting the liver, where PDAC frequently first spreads, is also given some consideration. Overall, the complementary measures are divided into three groups: dietary factors, nutraceutical agents and lifestyle. In turn, dietary factors are considered as general conditioners, multi-factorial foodstuffs and specific compounds. The general conditioners are alkalinity, low-glycemic index and low-cholesterol. The multi-factorial foodstuffs comprise red meat, fish, fruit/vegetables, dairy, honey and coffee. The available evidence for the beneficial effects of the specific dietary and nutraceutical agents was considered at four levels (in order of prominence): clinical trials, meta-analyses, in vivo tests and in vitro studies. Thus, 9 specific agents were identified (6 dietary and 3 nutraceutical) as acceptable for integration with gemcitabine chemotherapy, the first-line treatment for pancreatic cancer. The specific dietary agents were the following: Vitamins A, C, D and E, genistein and curcumin. As nutraceutical compounds, propolis, triptolide and cannabidiol were accepted. The 9 complementary agents were sub-grouped into two with reference to the main 'hallmarks of cancer'. Lifestyle factors covered obesity, diabetes, smoking, alcohol and exercise. An integrative treatment regimen was devised for the management of PDAC patients. This involved combining first-line gemcitabine chemotherapy with the two sub-groups of complementary agents alternately in weekly cycles. The review concludes that integrated management currently offers the best patient outcome. Opportunities to be investigated in the future include emerging modalities, precision medicine, the nerve input to tumors and, importantly, clinical trials.
Collapse
Affiliation(s)
- Valerie Jentzsch
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (V.J.); (J.A.A.D.)
- Business School, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - James A. A. Davis
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (V.J.); (J.A.A.D.)
| | - Mustafa B. A. Djamgoz
- Department of Life Sciences, Neuroscience Solutions to Cancer Research Group, Imperial College London, South Kensington Campus, London SW7 2AZ, UK; (V.J.); (J.A.A.D.)
- Biotechnology Research Centre, Cyprus International University, Haspolat, Nicosia, TRNC, Mersin 10, Turkey
| |
Collapse
|
12
|
Gromisch C, Qadan M, Machado MA, Liu K, Colson Y, Grinstaff MW. Pancreatic Adenocarcinoma: Unconventional Approaches for an Unconventional Disease. Cancer Res 2020; 80:3179-3192. [PMID: 32220831 PMCID: PMC7755309 DOI: 10.1158/0008-5472.can-19-2731] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/08/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022]
Abstract
This review highlights current treatments, limitations, and pitfalls in the management of pancreatic cancer and discusses current research in novel targets and drug development to overcome these clinical challenges. We begin with a review of the clinical landscape of pancreatic cancer, including genetic and environmental risk factors, as well as limitations in disease diagnosis and prevention. We next discuss current treatment paradigms for pancreatic cancer and the shortcomings of targeted therapy in this disease. Targeting major driver mutations in pancreatic cancer, such as dysregulation in the KRAS and TGFβ signaling pathways, have failed to improve survival outcomes compared with nontargeted chemotherapy; thus, we describe new advances in therapy such as Ras-binding pocket inhibitors. We then review next-generation approaches in nanomedicine and drug delivery, focusing on preclinical advancements in novel optical probes, antibodies, small-molecule agents, and nucleic acids to improve surgical outcomes in resectable disease, augment current therapies, expand druggable targets, and minimize morbidity. We conclude by summarizing progress in current research, identifying areas for future exploration in drug development and nanotechnology, and discussing future prospects for management of this disease.
Collapse
Affiliation(s)
- Christopher Gromisch
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Motaz Qadan
- Division of Surgical Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mariana Albuquerque Machado
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology and Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
| | - Yolonda Colson
- Division of Thoracic Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Mark W Grinstaff
- Departments of Pharmacology and Experimental Therapeutics, Biomedical Engineering, and Chemistry, Boston University, Boston, Massachusetts.
| |
Collapse
|
13
|
Immuno-OpenPET: a novel approach for early diagnosis and image-guided surgery for small resectable pancreatic cancer. Sci Rep 2020; 10:4143. [PMID: 32157106 PMCID: PMC7064510 DOI: 10.1038/s41598-020-61056-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/17/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer (PC) has a poor prognosis owing to difficulties in the diagnosis of resectable PC at early stages. Several clinical studies have indicated that the detection and surgery of small resectable PC (<1 cm) can significantly improve survival; however, imaging diagnosis and accurate resection of small PC remain challenging. Here, we report the feasibility of "immuno-OpenPET" as a novel approach enabling not only early diagnosis but also image-guided surgery, using a small (<1 cm) resectable PC orthotopic xenograft mouse model. For immuno-OpenPET, we utilized our original OpenPET system, which enables high-resolution positron emission tomography (PET) imaging with depth-of-interaction detectors, as well as real-time image-guided surgery, by arranging the detectors to create an open space for surgery and accelerating the image reconstruction process by graphics processing units. For immuno-OpenPET, 64Cu-labeled anti-epidermal growth factor receptor antibody cetuximab was intraperitoneally administered into mice. It clearly identified PC tumors ≥3 mm. In contrast, neither OpenPET with intravenous-administered 64Cu-cetuximab nor intraperitoneal/intravenous-administered 18F-FDG (a traditional PET probe) could detect PC in this model. Immuno-OpenPET-guided surgery accurately resected small PC in mice and achieved significantly prolonged survival. This technology could provide a novel diagnostic and therapeutic strategy for small resectable PC to improve patient survival.
Collapse
|
14
|
Zhou X, Gao W, Hua H, Ji Z. LncRNA-BLACAT1 Facilitates Proliferation, Migration and Aerobic Glycolysis of Pancreatic Cancer Cells by Repressing CDKN1C via EZH2-Induced H3K27me3. Front Oncol 2020; 10:539805. [PMID: 33072570 PMCID: PMC7538708 DOI: 10.3389/fonc.2020.539805] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE To investigate the role of lncRNA-BLACAT1 in promoting H3K27 trimethylation of CDKN1C gene by recruiting EZH2 to regulate CCNE on glycolysis and mitochondrial oxidative phosphorylation of pancreatic cancer (PC) cells. METHODS Following bioinformatic prediction, EZH2 and BLACAT1 in PC cells were interfered, and cells proliferation, migration and invasion in each group were detected. Western blotting detected the expression of key proteins of mitochondrial complex. The sub-cellular localization of BLACAT1 was tested, followed by testing the binding of CDKN1C and BLACAT1 with EZH2, followed by in vivo verification. RESULTS Based on bioinformatic prediction, EZH2 and BLACAT1 were highly expressed in PC, while CDKN1C was lowly expressed (all P < 0.05). Interference with EZH2 and BLACAT1 inhibited cell proliferation, migration and aerobic glycolysis, and promoted mitochondrial oxidative phosphorylation (all P < 0.05). BLACAT1 promoted H3K27 trimethylation of CDKN1C through recruiting EZH2 (all P < 0.05). In vivo results showed that BLACAT1 interference inhibited tumor formation (all P < 0.05). CONCLUSION Interference with BLACAT1 inhibits H3K27 trimethylation of CDKN1C gene by blocking EZH2 recruitment to promote CDKN1C expression and inhibit CCNE expression, thus suppressing PC cell proliferation, migration and aerobic glycolysis, and promoting mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Oncology, Linyi People’s Hospital, Linyi, China
| | - Wei Gao
- Department of Clinical Laboratory, Linyi People’s Hospital, Linyi, China
| | - Huanhuan Hua
- Department of Obstetrics and Gynecology, Kuitun Hospital of Yili Kazak Autonomous Prefecture, Yili Kazak Autonomous Prefecture, Xinjiang, China
| | - Zhimin Ji
- Department of Oncology, Linyi People’s Hospital, Linyi, China
- *Correspondence: Zhimin Ji,
| |
Collapse
|
15
|
Ferencz S, Reglodi D, Kaszas B, Bardosi A, Toth D, Vekony Z, Vicena V, Karadi O, Kelemen D. PACAP and PAC1 receptor expression in pancreatic ductal carcinoma. Oncol Lett 2019; 18:5725-5730. [PMID: 31788045 PMCID: PMC6865831 DOI: 10.3892/ol.2019.10971] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/12/2019] [Indexed: 12/26/2022] Open
Abstract
Pancreatic carcinoma is one of the most malignant diseases and is associated with a poor survival rate. Pituitary adenylate cyclase activating polypeptide (PACAP) is a neuropeptide that acts on three different G protein-coupled receptors: the specific PAC1 and the VPAC1/2 that also bind vasoactive intestinal peptide. PACAP is widely distributed in the body and has diverse physiological effects. Among other things, it acts as a trophic factor and influences proliferation and differentiation of several different cells both under normal circumstances and tumourous transformation. Changes of PACAP and its receptors have been shown in various tumour types. However, it is not known whether PACAP and its specific receptor are altered in pancreatic cancer. Perioperative data of patients with pancreas carcinoma was investigated over a five-year period. Histological results showed Grade 2 or Grade 3 adenocarcinoma in most cases. PACAP and PAC1 receptor expression were investigated by immunohistochemistry. Staining intensity of PAC1 receptor was strong in normal tissues both in the exocrine and endocrine parts of the pancreas, the receptor staining was markedly weaker in the adenocarcinoma. PACAP immunostaining was weak in the exocrine part and very strong in the islets and nerve elements in non-tumourous tissues. The PACAP immunostaining almost disappeared in the adenocarcinoma samples. Based on these findings a decrease or lack of the PAC1 receptor/PACAP signalling might have an influence on tumour growth and/or differentiation.
Collapse
Affiliation(s)
- Sandor Ferencz
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Dora Reglodi
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Balint Kaszas
- Department of Pathology, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Attila Bardosi
- Center for Histology, Cytology and Molecular Diagnostics, and Proteopath GmbH, Trier 54296, Germany
| | - Denes Toth
- Department of Forensic Medicine, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Zsofia Vekony
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Viktoria Vicena
- Department of Anatomy, MTA-PTE PACAP Research Group, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Oszkar Karadi
- Department of Oncology, University of Pécs, Medical School, Pécs 7622, Hungary
| | - Dezso Kelemen
- Department of Surgery, University of Pécs, Medical School, Pécs 7622, Hungary
| |
Collapse
|
16
|
Lee SW, Zhang Y, Jung M, Cruz N, Alas B, Commisso C. EGFR-Pak Signaling Selectively Regulates Glutamine Deprivation-Induced Macropinocytosis. Dev Cell 2019; 50:381-392.e5. [PMID: 31257175 DOI: 10.1016/j.devcel.2019.05.043] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 04/24/2019] [Accepted: 05/30/2019] [Indexed: 12/24/2022]
Abstract
Macropinocytosis has emerged as an important nutrient-scavenging pathway that supports tumor cell fitness. By internalizing extracellular protein and targeting it for lysosomal degradation, this endocytic pathway functions as an amino acid supply route, permitting tumor cell growth and survival despite the nutrient-poor conditions of the tumor microenvironment. Here, we provide evidence that a subset of pancreatic ductal adenocarcinoma (PDAC) tumors are wired to integrate contextual metabolic inputs to regulate macropinocytosis, dialing up or down this uptake pathway depending on nutrient availability. We find that regional depletion of amino acids coincides with increased levels of macropinocytosis and that the scarcity of glutamine uniquely drives this process. Mechanistically, this stimulation of macropinocytosis depends on the nutrient stress-induced potentiation of epidermal growth factor receptor signaling that, through the activation of Pak, controls the extent of macropinocytosis in these cells. These results provide a mechanistic understanding of how nutritional cues can control protein scavenging in PDAC tumors.
Collapse
Affiliation(s)
- Szu-Wei Lee
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yijuan Zhang
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Michael Jung
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Nathalia Cruz
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Basheer Alas
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Cosimo Commisso
- Tumor Initiation and Maintenance Program, NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Singh RR, Goldberg J, Varghese AM, Yu KH, Park W, O'Reilly EM. Genomic profiling in pancreatic ductal adenocarcinoma and a pathway towards therapy individualization: A scoping review. Cancer Treat Rev 2019; 75:27-38. [PMID: 30927677 PMCID: PMC6504563 DOI: 10.1016/j.ctrv.2019.03.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 12/11/2022]
Abstract
CONTEXT Pancreatic cancer (PDAC) is one of the most challenging cancers to treat with modest recent improvements in survival from new systemic therapies. There is growing interest in individualized therapy underpinned by somatic and germline genomic alterations. OBJECTIVE A systematic review of data on therapies targeting somatic and germline alterations, and their downstream pathways in PDAC. METHOD A systematic literature search was conducted using PRISMA guidelines to include relevant results published after January 1, 2008. RESULTS A total of 71 relevant studies were included. We identified 36 studies targeting the KRAS-pathway, the most common being with MEK-inhibitor therapy. Twenty-two studies were identified that evaluated platinum-based chemotherapy and PARP inhibitors in patients with deleterious mutations in DNA damage repair genes and have shown encouraging results. Immunotherapy has demonstrated activity in patients with mismatch repair deficiency/microsatellite instability. CONCLUSION Evidence from translational and clinical research presents an exciting platform for genomic targeted therapy in PDAC. Validity for targeting BRCA with platinum and PARP inhibitors and microsatellite instability with immune therapy has been established, nonetheless, evidence for targeting the common driver oncogenes is lacking and much work is needed. Of importance is identifying the subgroup of KRAS -wild type PDAC (approximately 5%) where there is enrichment for targetable opportunities.
Collapse
Affiliation(s)
- Ritu R Singh
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai St. Luke's and Mount Sinai West, New York, NY 10019, USA.
| | - Johanna Goldberg
- MSK Library, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Anna M Varghese
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; David M. Rubenstein Center for Pancreatic Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; David M. Rubenstein Center for Pancreatic Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; David M. Rubenstein Center for Pancreatic Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Weill Cornell Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; David M. Rubenstein Center for Pancreatic Research, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
18
|
Du J, He Y, Wu W, Li P, Chen Y, Hu Z, Han Y. Targeting EphA2 with miR-124 mediates Erlotinib resistance in K-RAS mutated pancreatic cancer. J Pharm Pharmacol 2019; 71:196-205. [PMID: 30604411 DOI: 10.1111/jphp.12941] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/19/2018] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Chemotheraputic drug resistance is a critical factor associated with the poor survival in advanced/metastatic pancreatic cancer (PC) patients. METHODS Human pancreatic cell lines Capan-1 and BXPC-3 were cultured with different concentrations of erlotinib (0, 10, 50, and 100 μm) for 48 h. The relative cell viability and apoptosis was detected using MTT assays and flow cytometry apoptosis analysis, respectively. Transfection of pcDNA-EphA2, si-EphA2 and miR-124 mimic/inhibitor was used to modulate the intracellular level of EphA2 and miR-124. The interaction between miR-124 and the 3'UTR of EphA2 was explored using dual luciferase reporter assay. KEY FINDINGS Compared with BXPC-3 cells, Capan-1 cells showed resistance to differential concentration treatment of erlotinib. The expression of EphA-2 was significantly increased and the expression of miR-124 was significantly decreased in Capan-1 cells. Overexpressing EphA2 induced resistance of BXPC-3 cells to erlotinib treatment. And EphA2 was identified as a novel target gene for miR-124. MiR-124 overexpression was able to sensitize the response of Capan-1 cells to erlotinib through inhibiting EphA2. Furthermore, both miR-124 overexpression and EphA2 inhibition sensitized Capan-1 cells to erlotinib in xenograft model. CONCLUSIONS Our study demonstrated that EphA2 rescued by miR-124 downregulation conferred the erlotinib resistance of PC cell Capan-1 with K-RAS mutation.
Collapse
Affiliation(s)
- Jing Du
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China.,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuanqiao He
- Department of Laboratory Animal Science, Nanchang University, Nanchang, Jiangxi, China
| | - Weiquan Wu
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China.,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Peng Li
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China.,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Youwei Chen
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China.,People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhiming Hu
- People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yong Han
- People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China.,Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Xu B, Zeng M, Zeng J, Feng J, Yu L. Meta-analysis of clinical trials comparing the efficacy and safety of liposomal cisplatin versus conventional nonliposomal cisplatin in nonsmall cell lung cancer (NSCLC) and squamous cell carcinoma of the head and neck (SCCHN). Medicine (Baltimore) 2018; 97:e13169. [PMID: 30431590 PMCID: PMC6257614 DOI: 10.1097/md.0000000000013169] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND While liposomal cisplatin has shown enhanced drug tolerability and higher targeting property as compared with the conventional cisplatin, the doubt remains whether lipoplatin could improve its anticancer efficacy. What's more, there is still no systematic evaluation of the safety profiles of lipoplatin comparing with original cisplatin. Thus, we performed a systematic literature search for randomized clinical trials directly comparing efficacy and safety of liposomal cisplatin versus its conventional nonliposomal cisplatin. METHODS The electronic search was conducted in PubMed, Embase, The Cochrane Library, and ClinicalTrials.gov from inception to February 10, 2018. The pooled odds ratio (OR) and 95% confidence intervals (CIs) of progressive disease (PD), partial response (PR), stable disease (SD), and adverse events (AEs) were obtained to assess the efficacy and safety. Heterogeneity was estimated using the I test (I > 50%, significant heterogeneity). RESULTS The search yielded 5 clinical trials that meet inclusion criteria, with a total of 523 patients. We found that the liposome encapsulated cisplatin was more clinical efficacious than cisplatin as assessed by PD rate (OR, 0.46; 95% CI, 0.28-0.74; P = .002), while subgroup analysis of the only nonsmall cell lung cancer (NSCLC) patients showed higher response rates in PR (OR, 0.46; 95% CI, 0.28-0.74; P = .002) and PD (OR, 0.46; 95% CI, 0.28-0.74; P = .002) simultaneously. In addition, the toxicity meta-analysis revealed lipoplatin was much less toxic than the original cisplatin, with respect to grade 3 to 4 neurotoxicity (OR, 0.18; 95% CI, 0.04-0.74; P = .02), grade 3 to 4 leukopenia (OR, 0.47; 95% CI, 0.26-0.85; P = .01), grade 3 to 4 neutropenia (OR, 0.26; 95% CI, 0.09-0.71; P = .009), grade 1 and 2 nausea/vomiting (OR, 0.50; 95% CI, 0.32-0.77; P = .002), and grade 3 and 4 asthenia (OR, 0.11; 95% CI, 0.03-0.42; P = .001). CONCLUSIONS This meta-analysis revealed that with both NSCLC and squamous cell carcinoma of the head and neck (SCCHN) patients, liposomal cisplatin-based chemotherapy offers significant advantages regarding the PD and reduced toxicities relative to conventional cisplatin.
Collapse
Affiliation(s)
- Bei Xu
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang
| | - Min Zeng
- Department of Pharmacy, Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiawei Zeng
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang
| | - Jiafu Feng
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang
| | - Lin Yu
- Department of Clinical Laboratory, Mianyang Central Hospital, Mianyang
| |
Collapse
|
20
|
Wang X, Sheng W, Wang Y, Li L, Li Y, Zhang S, Liu X, Chen S, Zhen Y. A Macropinocytosis-Intensifying Albumin Domain-Based scFv Antibody and Its Conjugate Directed against K-Ras Mutant Pancreatic Cancer. Mol Pharm 2018; 15:2403-2412. [PMID: 29757658 DOI: 10.1021/acs.molpharmaceut.8b00234] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Enhanced macropinocytosis has been found in K-Ras mutant pancreatic cancer cells, through which albumin can massively enter into the K-Ras-driven cancer cells, suggesting its role in serving as a macropinocytosis-intensifying drug delivery carrier. In the present study, a novel recombinant protein Fv-LDP-D3 and its reconstituted analogue Fv-LDP-D3-AE were designed and prepared. Fv is the fragment of an anti-EGFR antibody, D3 is the domain III of human serum albumin (HSA), LDP is the apoprotein of the antitumor antibiotic lidamycin (LDM), and AE is an extremely cytotoxic enediyne chromophore derived from LDM. As shown, the recombinant protein Fv-LDP-D3 presented intensive and selective binding capacity to pancreatic cancer cells and inhibited cell proliferation by blocking EGFR signaling. Moreover, Fv-LDP-D3 showed prominent tumor imaging in pancreatic carcinoma xenograft. The reconstituted, enediyne-integrated analogue Fv-LDP-D3-AE displayed highly potent cytotoxicity to pancreatic cancer cells through apoptosis induction and G2/M arrest. Fv-LDP-D3 and Fv-LDP-D3-AE markedly inhibited the tumor growth of the pancreatic carcinoma AsPC-1 xenograft. Study results indicated that the novel recombinant protein displays both EGFR-targeting and macropinocytosis-intensifying attributes, presenting a new format of scFv antibody that integrates with albumin domain III. It might be a feasible strategy to develop targeted drugs for K-Ras mutant pancreatic cancer.
Collapse
Affiliation(s)
- Xiaofei Wang
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Weijin Sheng
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Yangyang Wang
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Liang Li
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Yi Li
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Shenghua Zhang
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Xiujun Liu
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Shuzhen Chen
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| | - Yongsu Zhen
- Institute of Medicinal Biotechnology , Chinese Academy of Medical Sciences and Peking Union Medical College , No. 1 Tiantanxili , Beijing 100050 , China
| |
Collapse
|
21
|
Rzepecki AK, Cheng H, McLellan BN. Cutaneous toxicity as a predictive biomarker for clinical outcome in patients receiving anticancer therapy. J Am Acad Dermatol 2018; 79:545-555. [PMID: 29733938 DOI: 10.1016/j.jaad.2018.04.046] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/09/2018] [Accepted: 04/29/2018] [Indexed: 12/12/2022]
Abstract
The relationship between treatment outcome and cutaneous toxicity induced by anticancer therapy has gained attention in the past decade. In this article, we have provided an overview of the 3 main classes of anticancer agents-specifically, molecularly targeted kinase inhibitors, immune checkpoint inhibitors, and cytotoxic chemotherapeutics-and described the data evaluating the association between cutaneous toxicity induced by these agents and survival benefit. Although preliminary studies are promising with regard to the potential role of cutaneous toxicities as a surrogate biomarker of efficacy of treatment, larger prospective studies are needed to confirm this relationship. Dermatologists have a unique opportunity to collaborate with oncologists in the multidisciplinary treatment paradigm by helping to identify and manage these dermatologic events in patients with cancer. A heightened awareness of these toxicities is critical, as it can potentially allow recognition of the efficacy of anticancer therapy and may influence treatment decisions and patient outcomes.
Collapse
Affiliation(s)
- Alexandra K Rzepecki
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Michigan.
| | - Haiying Cheng
- Department of Oncology, Montefiore Medical Center, Bronx, New York
| | - Beth N McLellan
- Division of Dermatology, Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|