1
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
2
|
Alagic Z, Duran CV, Svensson-Marcial A, Koskinen SK. Contrast-enhanced photon-counting detector CT for discriminating local recurrence from postoperative changes after resection of pancreatic ductal adenocarcinoma. Eur Radiol Exp 2025; 9:26. [PMID: 39985649 PMCID: PMC11846822 DOI: 10.1186/s41747-025-00567-0] [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: 08/10/2024] [Accepted: 01/30/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND We evaluated the diagnostic capability of photon-counting detector computed tomography (PCD-CT) spectral variables in late arterial phase (LAP) and portal venous phase (PVP) to discriminate between local tumor recurrence (LTR) and postoperative changes (POC) after pancreatic ductal adenocarcinoma (PDAC) resection. METHODS Seventy-three consecutive PCD-CT scans in 73 patients with postoperative soft-tissue lesions (PSLs) were included, 42 with POC and 31 with LTR. Regions of interest were drawn in each PSL, and spectral variables were calculated: iodine concentration (IC), normalized IC (NIC), fat fraction, attenuation at 40, 70, and 90 keV, and slope of the spectral curve between 40-90 keV. Multivariable binary logistic regression models were constructed. Diagnostic performance was assessed for LAP and PVP using receiver operating characteristic analysis. RESULTS In LAP, all variables except fat fraction showed significant differences between LTR and POC (p ≤ 0.025). In PVP, all variables except NIC and fat fraction demonstrated significant differences between LTR and POC (p ≤ 0.005). Logistic regression analysis included NIC and 70 keV in the LAP-based model and IC and 90 keV in the PVP-based model. Both models achieved a higher area under the curve (AUC) than individual spectral variables in each phase. The LAP-based model achieved an AUC of 0.919 with 94% sensitivity, 84% specificity, and 87% accuracy, while the PVP-based model reached 0.820, 71%, 88%, and 81%, respectively. CONCLUSION Spectral variables from PCD-CT help distinguish between LTR and POC in LAP and PVP post-PDAC resection. Multivariable logistic regression improves diagnostic performance, especially in LAP. RELEVANCE STATEMENT Measuring normalized iodine concentration and attenuation at 70 keV in late arterial phase, or iodine concentration and attenuation at 90 keV in portal venous phase, and incorporating these values into a logistic regression model can help differentiate between local tumor recurrence and postoperative changes after pancreatic ductal adenocarcinoma resection. KEY POINTS Distinguishing recurrence from postoperative changes on CT after pancreatic ductal adenocarcinoma resection is challenging. PCD-CT spectral variable values differed significantly between local tumor recurrence (LTR) and postoperative changes (POC). Logistic regression of spectral variables can help distinguish LTR from POC. The late arterial phase-based model reached an AUC of 0.919 with 94% sensitivity and 84% specificity.
Collapse
Affiliation(s)
- Zlatan Alagic
- Department of Diagnostic Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden.
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 171 77, Stockholm, Sweden.
| | - Carlos Valls Duran
- Department of Diagnostic Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Anders Svensson-Marcial
- Department of Diagnostic Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Seppo K Koskinen
- Department of Diagnostic Radiology, Karolinska University Hospital, 171 76, Stockholm, Sweden
- Department of Clinical Science, Intervention, and Technology (CLINTEC), Karolinska Institutet, 171 77, Stockholm, Sweden
| |
Collapse
|
3
|
Carlomagno S, Setti C, Ortolani F, Sivori S. Pancreatic ductal adenocarcinoma microenvironment: Soluble factors and cancer associated fibroblasts as modulators of NK cell functions. Immunol Lett 2024; 269:106898. [PMID: 39019404 DOI: 10.1016/j.imlet.2024.106898] [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: 06/03/2024] [Accepted: 07/13/2024] [Indexed: 07/19/2024]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is the most frequent pancreatic cancer and represents one of the most aggressive human neoplasms. Typically identified at advance stage disease, most PDAC tumors are unresectable and resistant to standard therapies. The immunosuppressive microenvironment in PDAC impedes tumor control but a greater understanding of the complex stromal interactions within the tumor microenvironment (TME) and the development of strategies capable of restoring antitumor effector immune responses could be crucial to fight this aggressive tumor and its spread. Natural Killer (NK) cells play a crucial role in cancer immunosurveillance and represent an attractive target for immunotherapies, both as cell therapy and as a pharmaceutical target. This review describes some crucial components of the PDAC TME (collagens, soluble factors and fibroblasts) that can influence the presence, phenotype and function of NK cells in PDAC patients tumor tissue. This focused overview highlights the therapeutic relevance of dissecting the complex stromal composition to define new strategies for NK cell-based immunotherapies to improve the treatment of PDAC.
Collapse
Affiliation(s)
- Simona Carlomagno
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy.
| | - Chiara Setti
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy
| | - Fulvia Ortolani
- Department of Medicine (DMED), University of Udine, Piazzale Kolbe 4, Udine 33100, Italy
| | - Simona Sivori
- Department of Experimental Medicine (DIMES), University of Genoa, Via Leon Battista Alberti 2, Genoa 16132, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
4
|
Basu R, Kulkarni P, Swegan D, Duran-Ortiz S, Ahmad A, Caggiano LJ, Davis E, Walsh C, Brenya E, Koshal A, Brody R, Sandbhor U, Neggers SJCMM, Kopchick JJ. Growth Hormone Receptor Antagonist Markedly Improves Gemcitabine Response in a Mouse Xenograft Model of Human Pancreatic Cancer. Int J Mol Sci 2024; 25:7438. [PMID: 39000545 PMCID: PMC11242728 DOI: 10.3390/ijms25137438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/25/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Chemotherapy treatment against pancreatic ductal adenocarcinoma (PDAC) is thwarted by tumoral activation of multiple therapy resistance pathways. The growth hormone (GH)-GH receptor (GHR) pair is a covert driver of multimodal therapy resistance in cancer and is overexpressed in PDAC tumors, yet the therapeutic potential of targeting the same has not been explored. Here, we report that GHR expression is a negative prognostic factor in patients with PDAC. Combinations of gemcitabine with different GHR antagonists (GHRAs) markedly improve therapeutic outcomes in nude mice xenografts. Employing cultured cells, mouse xenografts, and analyses of the human PDAC transcriptome, we identified that attenuation of the multidrug transporter and epithelial-to-mesenchymal transition programs in the tumors underlie the observed augmentation of chemotherapy efficacy by GHRAs. Moreover, in human PDAC patients, GHR expression strongly correlates with a gene signature of tumor promotion and immune evasion, which corroborate with that in syngeneic tumors in wild-type vs. GH transgenic mice. Overall, we found that GH action in PDAC promoted a therapy-refractory gene signature in vivo, which can be effectively attenuated by GHR antagonism. Our results collectively present a proof of concept toward considering GHR antagonists to improve chemotherapeutic outcomes in the highly chemoresistant PDAC.
Collapse
MESH Headings
- Animals
- Gemcitabine
- Humans
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/pharmacology
- Deoxycytidine/therapeutic use
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/genetics
- Mice
- Xenograft Model Antitumor Assays
- Receptors, Somatotropin/metabolism
- Receptors, Somatotropin/antagonists & inhibitors
- Receptors, Somatotropin/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Cell Line, Tumor
- Mice, Nude
- Drug Resistance, Neoplasm/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Female
Collapse
Affiliation(s)
- Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Deborah Swegan
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Silvana Duran-Ortiz
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
| | - Arshad Ahmad
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Lydia J. Caggiano
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Honors Tutorial College, Ohio University, Athens, OH 45701, USA
| | - Emily Davis
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Christopher Walsh
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| | - Edward Brenya
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Adeel Koshal
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA;
| | - Rich Brody
- InfinixBio LLC, Columbus, OH 43212, USA; (R.B.); (U.S.)
| | - Uday Sandbhor
- InfinixBio LLC, Columbus, OH 43212, USA; (R.B.); (U.S.)
| | | | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (R.B.); (P.K.); (D.S.); (S.D.-O.); (A.A.); (L.J.C.); (E.D.); (C.W.); (E.B.)
- Diabetes Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
- Translational Biomedical Sciences Program, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
5
|
Jiménez DJ, Javed A, Rubio-Tomás T, Seye-Loum N, Barceló C. Clinical and Preclinical Targeting of Oncogenic Pathways in PDAC: Targeted Therapeutic Approaches for the Deadliest Cancer. Int J Mol Sci 2024; 25:2860. [PMID: 38474109 DOI: 10.3390/ijms25052860] [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: 12/21/2023] [Revised: 01/22/2024] [Accepted: 01/30/2024] [Indexed: 03/14/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death worldwide. It is commonly diagnosed in advanced stages and therapeutic interventions are typically constrained to systemic chemotherapy, which yields only modest clinical outcomes. In this review, we examine recent developments in targeted therapy tailored to address distinct molecular pathway alteration required for PDAC. Our review delineates the principal signaling pathways and molecular mechanisms implicated in the initiation and progression of PDAC. Subsequently, we provide an overview of prevailing guidelines, ongoing investigations, and prospective research trajectories related to targeted therapeutic interventions, drawing insights from randomized clinical trials and other pertinent studies. This review focus on a comprehensive examination of preclinical and clinical data substantiating the efficacy of these therapeutic modalities, emphasizing the potential of combinatorial regimens and novel therapies to enhance the quality of life for individuals afflicted with PDAC. Lastly, the review delves into the contemporary application and ongoing research endeavors concerning targeted therapy for PDAC. This synthesis serves to bridge the molecular elucidation of PDAC with its clinical implications, the evolution of innovative therapeutic strategies, and the changing landscape of treatment approaches.
Collapse
Affiliation(s)
- Diego J Jiménez
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Aadil Javed
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Teresa Rubio-Tomás
- School of Medicine, University of Crete, 70013 Herakleion, Crete, Greece
| | - Ndioba Seye-Loum
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| | - Carles Barceló
- Translational Pancreatic Cancer Oncogenesis Group, Health Research Institute of the Balearic Islands (IdISBa), Hospital Universitari Son Espases, 07120 Palma de Mallorca, Spain
| |
Collapse
|
6
|
Erreni M, Fumagalli MR, Zanini D, Candiello E, Tiberi G, Parente R, D’Anna R, Magrini E, Marchesi F, Cappello P, Doni A. Multiplexed Imaging Mass Cytometry Analysis in Preclinical Models of Pancreatic Cancer. Int J Mol Sci 2024; 25:1389. [PMID: 38338669 PMCID: PMC10855072 DOI: 10.3390/ijms25031389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers. PDAC is characterized by a complex tumor microenvironment (TME), that plays a pivotal role in disease progression and resistance to therapy. Investigating the spatial distribution and interaction of TME cells with the tumor is the basis for understanding the mechanisms underlying disease progression and represents a current challenge in PDAC research. Imaging mass cytometry (IMC) is the major multiplex imaging technology for the spatial analysis of tumor heterogeneity. However, there is a dearth of reports of multiplexed IMC panels for different preclinical mouse models, including pancreatic cancer. We addressed this gap by utilizing two preclinical models of PDAC: the genetically engineered, bearing KRAS-TP53 mutations in pancreatic cells, and the orthotopic, and developed a 28-marker panel for single-cell IMC analysis to assess the abundance, distribution and phenotypes of cells involved in PDAC progression and their reciprocal functional interactions. Herein, we provide an unprecedented definition of the distribution of TME cells in PDAC and compare the diversity between transplanted and genetic disease models. The results obtained represent an important and customizable tool for unraveling the complexities of PDAC and deciphering the mechanisms behind therapy resistance.
Collapse
Affiliation(s)
- Marco Erreni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
| | - Maria Rita Fumagalli
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Damiano Zanini
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Ermes Candiello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Giorgia Tiberi
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Raffaella Parente
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Raffaella D’Anna
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Elena Magrini
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Federica Marchesi
- IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milan, Italy
| | - Paola Cappello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Piazza Nizza 44b, 10126 Torino, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital -, via Manzoni 56, 20089 Rozzano, Milan, Italy
| |
Collapse
|
7
|
Hamel Z, Sanchez S, Standing D, Anant S. Role of STAT3 in pancreatic cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:20-34. [PMID: 38464736 PMCID: PMC10918236 DOI: 10.37349/etat.2024.00202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/22/2023] [Indexed: 03/12/2024] Open
Abstract
Pancreatic cancer remains a serious and deadly disease, impacting people globally. There remain prominent gaps in the current understanding of the disease, specifically regarding the role of the signal transducer and activator of transcription (STAT) family of proteins in pancreatic tumors. STAT proteins, particularly STAT3, play important roles in pancreatic cancer, especially pancreatic ductal adenocarcinoma (PDAC), which is the most prevalent histotype. The role of STAT3 across a continuum of molecular processes, such as PDAC tumorigenesis and progression, immune escape, drug resistance and stemness, and modulation of the tumor microenvironment (TME), are only a tip of the iceberg. In some ways, the role of STAT3 in PDAC may hold greater importance than that of oncogenic Kirsten rat sarcoma virus (KRAS). This makes STAT3 a highly attractive target for developing targeted therapies for the treatment of pancreatic cancer. In this review, the current knowledge of STAT3 in pancreatic cancer has been summarized, particularly relating to STAT3 activation in cancer cells, cells of the TME, and the state of targeting STAT3 in pre-clinical and clinical trials of PDAC.
Collapse
Affiliation(s)
- Zachary Hamel
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sierra Sanchez
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
8
|
Lee CM, Hwang Y, Jeong JW, Kim M, Lee J, Bae SJ, Ahn SG, Fang S. BRCA1 mutation promotes sprouting angiogenesis in inflammatory cancer-associated fibroblast of triple-negative breast cancer. Cell Death Discov 2024; 10:5. [PMID: 38182557 PMCID: PMC10770063 DOI: 10.1038/s41420-023-01768-5] [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: 10/25/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with inferior outcomes owing to its low treatment response and high invasiveness. Based on abundant cancer-associated fibroblasts (CAFs) and frequent mutation of breast cancer-associated 1 (BRCA1) in TNBC, the characteristics of CAFs in TNBC patients with BRCA1 mutation compared to wild-type were investigated using single-cell analysis. Intriguingly, we observed that characteristics of inflammatory CAFs (iCAFs) were enriched in patients with BRCA1 mutation compared to the wild-type. iCAFs in patients with BRCA1 mutation exhibited outgoing signals to endothelial cells (ECs) clusters, including chemokine (C-X-C motif) ligand (CXCL) and vascular endothelial growth factor (VEGF). During CXCL signaling, the atypical chemokine receptor 1 (ACKR1) mainly interacts with CXCL family members in tumor endothelial cells (TECs). ACKR1-high TECs also showed high expression levels of angiogenesis-related genes, such as ANGPT2, MMP1, and SELE, which might lead to EC migration. Furthermore, iCAFs showed VEGF signals for FLT1 and KDR in TECs, which showed high co-expression with tip cell marker genes, including ZEB1 and MAFF, involved in sprouting angiogenesis. Moreover, BRCA1 mutation patients with relatively abundant iCAFs and tip cell gene expression exhibited a limited response to neoadjuvant chemotherapy, including cisplatin and bevacizumab. Importantly, our study observed the intricate link between iCAFs-mediated angiogenesis and chemoresistance in TNBC with BRCA1 mutation.
Collapse
Affiliation(s)
- Chae Min Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yeseong Hwang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jae Woong Jeong
- Department of Medicine, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Minki Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Janghee Lee
- Department of Surgery, Sacred Heart Hospital, Hallym University, Dongtan, 18450, Republic of Korea
- Department of Medicine, Yonsei University Graduate School, Seoul, 03722, Republic of Korea
| | - Soong June Bae
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea
| | - Sung Gwe Ahn
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
- Institute for Breast Cancer Precision Medicine, Yonsei University College of Medicine, Seoul, 06273, Republic of Korea.
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
9
|
Behrens D, Pfohl U, Conrad T, Becker M, Brzezicha B, Büttner B, Wagner S, Hallas C, Lawlor R, Khazak V, Linnebacher M, Wartmann T, Fichtner I, Hoffmann J, Dahlmann M, Walther W. Establishment and Thorough Characterization of Xenograft (PDX) Models Derived from Patients with Pancreatic Cancer for Molecular Analyses and Chemosensitivity Testing. Cancers (Basel) 2023; 15:5753. [PMID: 38136299 PMCID: PMC10741928 DOI: 10.3390/cancers15245753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/24/2023] Open
Abstract
Patient-derived xenograft (PDX) tumor models are essential for identifying new biomarkers, signaling pathways and novel targets, to better define key factors of therapy response and resistance mechanisms. Therefore, this study aimed at establishing pancreas carcinoma (PC) PDX models with thorough molecular characterization, and the identification of signatures defining responsiveness toward drug treatment. In total, 45 PC-PDXs were generated from 120 patient tumor specimens and the identity of PDX and corresponding patient tumors was validated. The majority of engrafted PDX models represent ductal adenocarcinomas (PDAC). The PDX growth characteristics were assessed, with great variations in doubling times (4 to 32 days). The mutational analyses revealed an individual mutational profile of the PDXs, predominantly showing alterations in the genes encoding KRAS, TP53, FAT1, KMT2D, MUC4, RNF213, ATR, MUC16, GNAS, RANBP2 and CDKN2A. Sensitivity of PDX toward standard of care (SoC) drugs gemcitabine, 5-fluorouracil, oxaliplatin and abraxane, and combinations thereof, revealed PDX models with sensitivity and resistance toward these treatments. We performed correlation analyses of drug sensitivity of these PDX models and their molecular profile to identify signatures for response and resistance. This study strongly supports the importance and value of PDX models for improvement in therapies of PC.
Collapse
Affiliation(s)
- Diana Behrens
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Ulrike Pfohl
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Theresia Conrad
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Michael Becker
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Bernadette Brzezicha
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Britta Büttner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Silvia Wagner
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, 72076 Tübingen, Germany
| | - Cora Hallas
- Institut für Hämatopathologie, Fangdieckstr. 75, 22547 Hamburg, Germany
| | - Rita Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Piazzale A. Scuro 10, 37134 Verona, Italy
| | | | - Michael Linnebacher
- Clinic of General Surgery, Molecular Oncology and Immunotherapy, University Medical Center Rostock, 18057 Rostock, Germany
| | - Thomas Wartmann
- University Clinic for General, Visceral, Vascular and Transplantation Surgery, Faculty of Medicine, Otto-von-Guericke-University, 39120 Magdeburg, Germany
| | - Iduna Fichtner
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Jens Hoffmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Mathias Dahlmann
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
| | - Wolfgang Walther
- Experimental Pharmacology and Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany (M.D.)
- Experimental and Clinical Research Center, Charité Universitätsmedizin Berlin, Lindenberger Weg 80, 13125 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| |
Collapse
|
10
|
Roos-Mattila M, Kaprio T, Mustonen H, Hagström J, Saharinen P, Haglund C, Seppänen H. The possible dual role of Ang-2 in the prognosis of pancreatic cancer. Sci Rep 2023; 13:18725. [PMID: 37907568 PMCID: PMC10618172 DOI: 10.1038/s41598-023-45194-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/17/2023] [Indexed: 11/02/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) features a dense desmoplastic stroma, which raises the intratumoral interstitial pressure leading to vascular collapse and hypoxia, inducing angiogenesis. Vascular growth factors, such as vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang-2), increase in PDAC. A high VEGF and a high circulating Ang-2 associate with shorter survival in PDAC. In addition to the circulatory Ang-2, PDAC endothelial and epithelial cells express Ang-2. No correlation between tumor epithelial nor endothelial cell Ang-2 expression and survival has been published. We aimed to examine Ang-2 expression and survival. This study comprised PDAC surgical patients at Helsinki University Hospital in 2000-2013. Ang-2 immunohistochemistry staining was completed on 168 PDAC patient samples. Circulating Ang-2 levels were measured using ELISA in the sera of 196 patients. Ang-2 levels were assessed against clinical data and patient outcomes. A low tumor epithelial Ang-2 expression predicted shorter disease-specific survival (DSS) compared with a high expression (p = 0.003). A high serum Ang-2 associated with shorter DSS compared with a low circulating Ang-2 (p = 0.016). Ang-2 seemingly plays a dual role in PDAC survival. Further studies are needed to determine the mechanisms causing tumor cell Ang-2 expression and its positive association with survival.
Collapse
Affiliation(s)
- Matilda Roos-Mattila
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Tuomas Kaprio
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland.
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
- Department of Pathology, Haartmaninkatu 3 (PB 21), University of Helsinki, 00014, Helsinki, Finland.
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jaana Hagström
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Oral Pathology and Radiology, University of Turku, Turku, Finland
| | - Pipsa Saharinen
- Wihuri Research Institute, Biomedicum Helsinki, University of Helsinki, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- Department of Surgery, Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- iCAN, Digital Cancer Precision Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
11
|
Natu J, Nagaraju GP. Gemcitabine effects on tumor microenvironment of pancreatic ductal adenocarcinoma: Special focus on resistance mechanisms and metronomic therapies. Cancer Lett 2023; 573:216382. [PMID: 37666293 DOI: 10.1016/j.canlet.2023.216382] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/26/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is considered one of the deadliest malignancies, with dismal survival rates and extremely prevalent chemoresistance. Gemcitabine is one of the primary treatments used in treating PDACs, but its benefits are limited due to chemoresistance, which could be attributed to interactions between the tumor microenvironment (TME) and intracellular processes. In preclinical models, certain schedules of administration of gemcitabine modulate the TME in a manner that does not promote resistance. Metronomic therapy constitutes a promising strategy to overcome some barriers associated with current PDAC treatments. This review will focus on gemcitabine's mechanism in treating PDAC, combination therapies, gemcitabine's interactions with the TME, and gemcitabine in metronomic therapies.
Collapse
Affiliation(s)
- Jay Natu
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA.
| |
Collapse
|
12
|
Karam M, Auclair C. Sphingosine-1-Phosphate as Lung and Cardiac Vasculature Protecting Agent in SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:13088. [PMID: 37685894 PMCID: PMC10488186 DOI: 10.3390/ijms241713088] [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: 06/25/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may cause severe respiratory illness with high mortality. SARS-CoV-2 infection results in a massive inflammatory cell infiltration into the infected lungs accompanied by excessive pro-inflammatory cytokine production. The lung histology of dead patients shows that some areas are severely emphysematous, with enormously dilated blood vessels and micro-thromboses. The inappropriate inflammatory response damaging the pulmonary interstitial arteriolar walls suggests that the respiratory distress may come in a large part from lung vasculature injuries. It has been recently observed that low plasmatic sphingosine-1-phosphate (S1P) is a marker of a worse prognosis of clinical outcome in severe coronavirus disease (COVID) patients. S1P is an angiogenic molecule displaying anti-inflammatory and anti-apoptotic properties, that promote intercellular interactions between endothelial cells and pericytes resulting in the stabilization of arteries and capillaries. In this context, it can be hypothesized that the benefit of a normal S1P level is due to its protective effect on lung vasculature functionality. This paper provides evidence supporting this concept, opening the way for the design of a pharmacological approach involving the use of an S1P lyase inhibitor to increase the S1P level that in turn will rescue the lung vasculature functionality.
Collapse
Affiliation(s)
| | - Christian Auclair
- AC BioTech, Villejuif Biopark, Cancer Campus, 1 mail du Professeur Georges Mathé, 94800 Villejuif, France;
| |
Collapse
|
13
|
Di Molfetta D, Cannone S, Greco MR, Caroppo R, Piccapane F, Carvalho TMA, Altamura C, Saltarella I, Tavares Valente D, Desaphy JF, Reshkin SJ, Cardone RA. ECM Composition Differentially Regulates Intracellular and Extracellular pH in Normal and Cancer Pancreatic Duct Epithelial Cells. Int J Mol Sci 2023; 24:10632. [PMID: 37445810 DOI: 10.3390/ijms241310632] [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: 04/07/2023] [Revised: 06/07/2023] [Accepted: 06/17/2023] [Indexed: 07/15/2023] Open
Abstract
Intracellular pH (pHi) regulation is a challenge for the exocrine pancreas, where the luminal secretion of bicarbonate-rich fluid is accompanied by interstitial flows of acid. This acid-base transport requires a plethora of ion transporters, including bicarbonate transporters and the Na+/H+ exchanger isoform 1 (NHE1), which are dysregulated in Pancreatic Ductal Adenocarcinoma (PDAC). PDAC progression is favored by a Collagen-I rich extracellular matrix (ECM) which exacerbates the physiological interstitial acidosis. In organotypic cultures of normal human pancreatic cells (HPDE), parenchymal cancer cells (CPCs) and cancer stem cells (CSCs) growing on matrices reproducing ECM changes during progression, we studied resting pHi, the pHi response to fluxes of NaHCO3 and acidosis and the role of NHE1 in pHi regulation. Our findings show that: (i) on the physiological ECM, HPDE cells have the most alkaline pHi, followed by CSCs and CPCs, while a Collagen I-rich ECM reverses the acid-base balance in cancer cells compared to normal cells; (ii) both resting pHi and pHi recovery from an acid load are reduced by extracellular NaHCO3, especially in HPDE cells on a normal ECM; (iii) cancer cell NHE1 activity is less affected by NaHCO3. We conclude that ECM composition and the fluctuations of pHe cooperate to predispose pHi homeostasis towards the presence of NaHCO3 gradients similar to that expected in the tumor.
Collapse
Affiliation(s)
- Daria Di Molfetta
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Stefania Cannone
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Rosa Caroppo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Francesca Piccapane
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | | | - Concetta Altamura
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ilaria Saltarella
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Diana Tavares Valente
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Jean Francois Desaphy
- Department of Biomedical Sciences and Human Oncology, School of Medicine, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Stephan J Reshkin
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70126 Bari, Italy
| |
Collapse
|
14
|
Qiu H, Su N, Wang J, Yan S, Li J. Quantitative proteomics analysis in small cell carcinoma of cervix reveals novel therapeutic targets. Clin Proteomics 2023; 20:18. [PMID: 37031178 PMCID: PMC10082492 DOI: 10.1186/s12014-023-09408-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/02/2023] [Indexed: 04/10/2023] Open
Abstract
BACKGROUND As a rare pathologic subtype, small cell carcinoma of the cervix (SCCC) is characterized by extensive aggressiveness and resistance to current therapies. To date, our knowledge of SCCC origin and progression is limited and sometimes even controversial. Herein, we explored the whole-protein expression profiles in a panel of SCCC cases, aiming to provide more evidence for the precise diagnosis and targeting therapy. METHODS Eighteen SCCC samples and six matched normal cervix tissues were collected from January 2013 to December 2017. Data independent acquisition mass spectrometry (DIA) was performed to discriminate the different proteins (DEPs) associated with SCCC. The expression of CDN2A and SYP in corresponding SCCC tissues was verified using immunohistochemistry. GO and KEGG enrichment analyses were used to identify the key DEPs related to SCCC development and tumor recurrence. RESULTS As a result, 1311 DEPs were identified in SCCC tissues (780 up-regulated and 531 down-regulated). In up-regulated DEPs, both GO analysis and KEGG analysis showed the most enriched were related to DNA replication (including nuclear DNA replication, DNA-dependent DNA replication, and cell cycle DNA replication), indicating the prosperous proliferation in SCCC. As for the down-regulated DEPs, GO analysis showed that the most enriched functions were associated with extracellular matrix collagen-containing extracellular matrix. KEGG analysis revealed that the DEPs were enriched in Complement and coagulation cascades, proteoglycans in cancer, and focal adhesion-related pathways. Down-regulation of these proteins could enhance the mobility of cancer cells and establish a favorable microenvironment for tumor metastasis, which might be accounted for the frequent local and distant metastasis in SCCC. Surprisingly, the blood vessels and circulatory system exhibit a down-regulation in SCCC, which might be partly responsible for its resistance to anti-angiogenic regimens. In the stratification analysis of early-stage tumors, a group of enzymes involved in the cancer metabolism was discriminated in these recurrence cases. CONCLUSIONS Using quantitative proteomics analysis, we first reported the whole-protein expression profiles in SCCC. Significant alterations were found in proteins associated with the enhancement of DNA replication and cellular motility. Besides the association with mitosis, a unique metabolic feature was detected in cases with tumor recurrence. These findings provided novel targets for disease surveillance and treatments, which warranted further validation in the future.
Collapse
Affiliation(s)
- Haifeng Qiu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Provincial Medical Key Laboratory for Gynecologic Malignancies Prevention and Treatment, Zhengzhou, Henan, China
- Zhengzhou Key Laboratory for Gynecologic Malignancies Prevention and Treatment, Zhengzhou, Henan, China
| | - Ning Su
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Shuping Yan
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, East Jianshe Road, Erqi District, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
15
|
Liu X, Iovanna J, Santofimia-Castaño P. Stroma-targeting strategies in pancreatic cancer: a double-edged sword. J Physiol Biochem 2023; 79:213-222. [PMID: 36580230 DOI: 10.1007/s13105-022-00941-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 12/19/2022] [Indexed: 12/30/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a type of cancer with limited treatment options and terrible long-term survival, and it is expected to become the second leading cause of cancer-related death by 2030. One reason why this cancer is so aggressive and resistant is the formation of dense stroma that surrounds the neoplastic epithelium, which promotes tumor progression, invasion, metastasis, and resistance. The three major components of PDAC stroma are extracellular matrix (ECM), cancer-associated fibroblasts (CAFs), and vasculature. The dense ECM acts as a natural physical barrier, impeding drug penetration to PDAC tumor cells. Consequently, the method that combines stroma-targeting with anticancer therapy may be a viable alternative for increasing drug penetration. Additionally, blood vessels are key entities of the tumor stroma, serving as a pathway for nutrition as well as the only way for chemical medicines and immune cells to act. Finally, PDAC CAFs and tumor cells have crosstalk effects in the tumor microenvironment, where they are responsible for enhanced matrix deposition. In this review, we aim to provide an overview of our current comprehension of the three key components of PDAC stroma and the new promising therapeutic targets for PDAC.
Collapse
Affiliation(s)
- Xi Liu
- Centre de Recherche en Cancérologie de Marseille (CRCM), UMR 7258, INSERM U1068, CNRS, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France
| | - Juan Iovanna
- Centre de Recherche en Cancérologie de Marseille (CRCM), UMR 7258, INSERM U1068, CNRS, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France
| | - Patricia Santofimia-Castaño
- Centre de Recherche en Cancérologie de Marseille (CRCM), UMR 7258, INSERM U1068, CNRS, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique Et Technologique de Luminy, 163 Avenue de Luminy, 13288, Marseille, France.
| |
Collapse
|
16
|
Gupta P, Velliou EG. A Step-by-Step Methodological Guide for Developing Zonal Multicellular Scaffold-Based Pancreatic Cancer Models. Methods Mol Biol 2023; 2645:221-229. [PMID: 37202622 DOI: 10.1007/978-1-0716-3056-3_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The tumor microenvironment (TME), a complex heterogeneous mixture of various cellular, physical, and biochemical components and signals, is a major player in the process of tumor growth and its response to therapeutic methods. In vitro 2D monocellular cancer models are unable to mimic the complex in vivo characteristics of cancer TME involving cellular heterogeneity, presence of extracellular matrix (ECM) proteins, as well as spatial orientation and organization of different cell types forming the TME. In vivo animal-based studies have ethical concerns, are expensive and time-consuming, and involve models of non-human species. In vitro 3D models are capable of tiding over several issues associated with both 2D in vitro and in vivo animal models. We have recently developed a novel zonal multicellular 3D in vitro model for pancreatic cancer involving cancer cells, endothelial cells, and pancreatic stellate cells. Our model (i) can provide long-term culture (up to 4 weeks), (ii) can control the ECM biochemical configuration in a cell specific manner, (iii) shows large amounts of collagen secretion by the stellate cells mimicking desmoplasia, and (iv) expresses cell-specific markers throughout the whole culture period. This chapter describes the experimental methodology to form our hybrid multicellular 3D model for pancreatic ductal adenocarcinoma, including the immunofluorescence staining on the cell culture.
Collapse
Affiliation(s)
- Priyanka Gupta
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, UK
| | - Eirini G Velliou
- Centre for 3D models of Health and Disease, Division of Surgery and Interventional Science, University College London, London, UK.
| |
Collapse
|
17
|
Metabolic Pathways as a Novel Landscape in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14153799. [PMID: 35954462 PMCID: PMC9367608 DOI: 10.3390/cancers14153799] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Metabolism plays a fundamental role in both human physiology and pathology, including pancreatic ductal adenocarcinoma (PDAC) and other tumors. Anabolic and catabolic processes do not only have energetic implications but are tightly associated with other cellular activities, such as DNA duplication, redox reactions, and cell homeostasis. PDAC displays a marked metabolic phenotype and the observed reduction in tumor growth induced by calorie restriction with in vivo models supports the crucial role of metabolism in this cancer type. The aggressiveness of PDAC might, therefore, be reduced by interventions on bioenergetic circuits. In this review, we describe the main metabolic mechanisms involved in PDAC growth and the biological features that may favor its onset and progression within an immunometabolic context. We also discuss the need to bridge the gap between basic research and clinical practice in order to offer alternative therapeutic approaches for PDAC patients in the more immediate future.
Collapse
|
18
|
Scarpellino G, Genova T, Quarta E, Distasi C, Dionisi M, Fiorio Pla A, Munaron L. P2X Purinergic Receptors Are Multisensory Detectors for Micro-Environmental Stimuli That Control Migration of Tumoral Endothelium. Cancers (Basel) 2022; 14:2743. [PMID: 35681724 PMCID: PMC9179260 DOI: 10.3390/cancers14112743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
The tumoral microenvironment often displays peculiar features, including accumulation of extracellular ATP, hypoxia, low pH-acidosis, as well as an imbalance in zinc (Zn2+) and calcium (Ca2+). We previously reported the ability of some purinergic agonists to exert an anti-migratory activity on tumor-derived human endothelial cells (TEC) only when applied at a high concentration. They also trigger calcium signals associated with release from intracellular stores and calcium entry from the external medium. Here, we provide evidence that high concentrations of BzATP (100 µM), a potent agonist of P2X receptors, decrease migration in TEC from different tumors, but not in normal microvascular ECs (HMEC). The same agonist evokes a calcium increase in TEC from the breast and kidney, as well as in HMEC, but not in TEC from the prostate, suggesting that the intracellular pathways responsible for the P2X-induced impairment of TEC migration could vary among different tumors. The calcium signal is mainly due to a long-lasting calcium entry from outside and is strictly dependent on the presence of the receptor occupancy. Low pH, as well as high extracellular Zn2+ and Ca2+, interfere with the response, a distinctive feature typically found in some P2X purinergic receptors. This study reveals that a BzATP-sensitive pathway impairs the migration of endothelial cells from different tumors through mechanisms finely tuned by environmental factors.
Collapse
Affiliation(s)
- Giorgia Scarpellino
- Department of Life Sciences & Systems Biology, University of Torino, 10123 Torino, Italy; (G.S.); (T.G.); (E.Q.); (A.F.P.)
| | - Tullio Genova
- Department of Life Sciences & Systems Biology, University of Torino, 10123 Torino, Italy; (G.S.); (T.G.); (E.Q.); (A.F.P.)
| | - Elisa Quarta
- Department of Life Sciences & Systems Biology, University of Torino, 10123 Torino, Italy; (G.S.); (T.G.); (E.Q.); (A.F.P.)
| | - Carla Distasi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.D.); (M.D.)
| | - Marianna Dionisi
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, 28100 Novara, Italy; (C.D.); (M.D.)
| | - Alessandra Fiorio Pla
- Department of Life Sciences & Systems Biology, University of Torino, 10123 Torino, Italy; (G.S.); (T.G.); (E.Q.); (A.F.P.)
| | - Luca Munaron
- Department of Life Sciences & Systems Biology, University of Torino, 10123 Torino, Italy; (G.S.); (T.G.); (E.Q.); (A.F.P.)
| |
Collapse
|
19
|
Koltai T, Reshkin SJ, Carvalho TMA, Di Molfetta D, Greco MR, Alfarouk KO, Cardone RA. Resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma: A Physiopathologic and Pharmacologic Review. Cancers (Basel) 2022; 14:2486. [PMID: 35626089 PMCID: PMC9139729 DOI: 10.3390/cancers14102486] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive tumor with a poor prognosis and inadequate response to treatment. Many factors contribute to this therapeutic failure: lack of symptoms until the tumor reaches an advanced stage, leading to late diagnosis; early lymphatic and hematic spread; advanced age of patients; important development of a pro-tumoral and hyperfibrotic stroma; high genetic and metabolic heterogeneity; poor vascular supply; a highly acidic matrix; extreme hypoxia; and early development of resistance to the available therapeutic options. In most cases, the disease is silent for a long time, andwhen it does become symptomatic, it is too late for ablative surgery; this is one of the major reasons explaining the short survival associated with the disease. Even when surgery is possible, relapsesare frequent, andthe causes of this devastating picture are the low efficacy ofand early resistance to all known chemotherapeutic treatments. Thus, it is imperative to analyze the roots of this resistance in order to improve the benefits of therapy. PDAC chemoresistance is the final product of different, but to some extent, interconnected factors. Surgery, being the most adequate treatment for pancreatic cancer and the only one that in a few selected cases can achieve longer survival, is only possible in less than 20% of patients. Thus, the treatment burden relies on chemotherapy in mostcases. While the FOLFIRINOX scheme has a slightly longer overall survival, it also produces many more adverse eventsso that gemcitabine is still considered the first choice for treatment, especially in combination with other compounds/agents. This review discusses the multiple causes of gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Khalid Omer Alfarouk
- Zamzam Research Center, Zamzam University College, Khartoum 11123, Sudan;
- Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| |
Collapse
|
20
|
Dai H, Abdullah R, Wu X, Li F, Ma Y, Lu A, Zhang G. Pancreatic Cancer: Nucleic Acid Drug Discovery and Targeted Therapy. Front Cell Dev Biol 2022; 10:855474. [PMID: 35652096 PMCID: PMC9149368 DOI: 10.3389/fcell.2022.855474] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 04/07/2022] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal cancers with an almost 10% 5-year survival rate. Because PC is implicated in high heterogeneity, desmoplastic tumor-microenvironment, and inefficient drug-penetration, the chemotherapeutic strategy currently recommended for the treatment of PC has limited clinical benefit. Nucleic acid-based targeting therapies have become strong competitors in the realm of drug discovery and targeted therapy. A vast evidence has demonstrated that antibody-based or alternatively aptamer-based strategy largely contributed to the elevated drug accumulation in tumors with reduced systematic cytotoxicity. This review describes the advanced progress of antisense oligonucleotides (ASOs), small interfering RNAs (siRNAs), microRNAs (miRNAs), messenger RNA (mRNAs), and aptamer-drug conjugates (ApDCs) in the treatment of PC, revealing the bright application and development direction in PC therapy.
Collapse
Affiliation(s)
- Hong Dai
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Razack Abdullah
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute for the Advancement of Chinese medicine (IACM) .Ltd, Shatin, Hong Kong SAR, China
| | - Xiaoqiu Wu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Fangfei Li
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Yuan Ma
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Aiping Lu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen, China
| |
Collapse
|
21
|
Liu W, Yin B, Liang ZH, Yu Y, Lu N. Computed tomography perfusion imaging evaluation of angiogenesis in patients with pancreatic adenocarcinoma. World J Clin Cases 2022; 10:2393-2403. [PMID: 35434057 PMCID: PMC8968604 DOI: 10.12998/wjcc.v10.i8.2393] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/30/2021] [Accepted: 02/16/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma is one of the most common malignant tumors of the digestive system. More than 80% of patients with pancreatic adenocarcinoma are not diagnosed until late stage and have distant or local metastases.
AIM To investigate the value of computed tomography (CT) perfusion imaging in the evaluation of angiogenesis in pancreatic adenocarcinoma patients.
METHODS This is a retrospective cohort study. Patients with pancreatic adenocarcinoma and volunteers without pancreatic diseases underwent CT perfusion imaging from December 2014 to August 2017 in Huashan Hospital, Fudan University Shanghai, China.
RESULTS A total number of 35 pancreatic adenocarcinoma patients and 33 volunteers were enrolled. The relative blood flow (rBF), and relative blood volume (rBV) were significantly lower in patients with pancreatic adenocarcinoma than in the control group (P < 0.05). Conversely, the relative permeability in patients with pancreatic adenocarcinoma was significantly higher than that in controls (P < 0.05). In addition, rBF, rBV, and the vascular maturity index (VMI) were significantly lower in grade III-IV pancreatic adenocarcinoma than in grade I-II pancreatic adenocarcinoma (P < 0.05). Vascular endothelial growth factor (VEGF), CD105-MVD, CD34-MVD, and angiogenesis rate (AR) were significantly higher in grade III-IV pancreatic adenocarcinoma than in grade I-II pancreatic adenocarcinoma (P < 0.05). Significant correlations between rBF and VEGF, CD105-MVD, AR, and VMI (P < 0.01) were observed. Moreover, the levels of rBV were statistically significantly correlated with those of VEGF, CD105-MVD, CD34-MVD, and VMI (P < 0.01).
CONCLUSION Perfusion CT imaging may be an appropriate approach for quantitative assessment of tumor angiogenesis in pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Wen Liu
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200000, China
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai 200000, China
| | - Zong-Hui Liang
- Department of Radiology, Shanghai Jing’an District Central Hospital, Huashan Hospital Jing’an Branch, Fudan University, Shanghai 200000, China
| | - Yang Yu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200000, China
| | - Na Lu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200000, China
- Department of Radiology, Huashan Hospital North, Fudan University, Shanghai 200000, China
| |
Collapse
|
22
|
Is Sphingosine-1-Phosphate a Regulator of Tumor Vascular Functionality? Cancers (Basel) 2022; 14:cancers14051302. [PMID: 35267610 PMCID: PMC8909747 DOI: 10.3390/cancers14051302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Despite substantial theoretical and experimental support for using vascular normalization as cancer therapy, effectively achieving this strategy in the clinic remains complex. In the present paper, we propose a novel potential approach for the induction of tumor vascular normalization, reduction of hypoxia, and improvement of conventional treatment in cancer patients. This approach consists of the pharmacological modulation of a patient’s plasma S1P levels which through a PDGF signaling can enhance tumor vasculature functionality and reduce hypoxia. This approach is proposed following a clinical observation in pancreatic adenocarcinoma patients and pre-clinical data in different in vivo tumor models, and is supported by a review of the literature describing the biological role of S1P in vascular functionality regulation. Abstract Increasing evidence indicates that tumor vasculature normalization could be an appropriate strategy to increase therapies’ efficacy in solid tumors by decreasing hypoxia and improving drug delivery. We searched for a novel approach that reduces hypoxia and enhances chemotherapy efficacy in pancreatic adenocarcinoma which is characterized by disrupted blood vasculature associated with poor patient survival. Clinical significance of plasma levels of the angiogenic lipid sphingosine-1-phosphate (S1P) was assessed at baseline in 175 patients. High plasma S1P concentration was found to be a favorable prognostic/predictive marker in advanced/metastatic pancreatic adenocarcinoma patients treated by gemcitabine alone but not in patients receiving a combination gemcitabine and PDGFR-inhibitor. In pancreatic adenocarcinoma PDX models, oral administration of an S1P lyase inhibitor (LX2931) significantly increased plasma S1P levels, decreased tumor expression of the hypoxia marker (CA IX), and enhanced chemotherapy efficacy when combined with gemcitabine treatment. The direct effect of S1P on tumor oxygenation was assessed by administration of S1P onto tumor-grafted CAM model and measuring intra-tumoral pO2 using a tissue oxygen monitor. S1P increased pO2 in a tumor-CAM model. Thus, increasing plasma S1P is a promising strategy to decrease tumor hypoxia and enhance therapy efficacy in solid tumors. S1P may act as a tumor vasculature normalizer.
Collapse
|
23
|
Janjua KA, Shahzad R, Shehzad A. Development of Novel Cancer Biomarkers for Diagnosis and Prognosis. CANCER BIOMARKERS IN DIAGNOSIS AND THERAPEUTICS 2022:277-343. [DOI: 10.1007/978-981-16-5759-7_11] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
24
|
Chen K, Wang Q, Liu X, Wang F, Yang Y, Tian X. Hypoxic pancreatic cancer derived exosomal miR-30b-5p promotes tumor angiogenesis by inhibiting GJA1 expression. Int J Biol Sci 2022; 18:1220-1237. [PMID: 35173549 PMCID: PMC8771853 DOI: 10.7150/ijbs.67675] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 12/25/2021] [Indexed: 11/05/2022] Open
Abstract
Purpose: Most patients with pancreatic ductal adenocarcinoma (PDAC) have vascular invasion and metastasis, leading to low surgical resection rate and dismal prognosis. Tumor angiogenesis is related to vascular invasion and metastasis. However, anti-angiogenesis therapeutic effects in PDAC are limited. Therefore, it is imperative to explore molecular mechanism of angiogenesis in PDAC. Experimental Design: scRNA-seq data were utilized to delineatetranscriptional profiles of endothelial cells in PDAC. The in vitro and vivo angiogenesis models were used to explore the role of PDAC derived exosomes under hypoxic condition in tumor angiogenesis. Results: Endothelial cells in PDAC had distinct gene expression profiles compared with normal pancreas. The marker genes of endothelial cells in PDAC were enriched for hypoxia and angiogenesis. MiR-30b-5p were significantly enriched in hypoxic PDAC cells derived exosomes, which could be transferred to HUVEC, resulting in the upregulation of miR-30b-5p. Hypoxic PDAC cells derived exosomes could promote tube formation and endothelial cells migration via miR-30b-5p mediated downregulation of gap junction protein GJA1. Moreover, hypoxic PDAC cells derived exosomes increased new microvascular density in vivo. Patients with PDAC had higher levels of total miR-30b-5p and exosomal miR-30b-5p in peripheral blood plasma than healthy subjects. In addition, there were significant correlations for the levels of total miR-30b-5p or exosomal miR-30b-5p between peripheral blood plasma and portal vein plasma. Conclusions: Hypoxic PDAC cells derived exosomal miR-30b-5p promoted angiogenesis by inhibiting GJA1, and miR-30b-5p was a potential diagnostic marker for PDAC.
Collapse
Affiliation(s)
- Kai Chen
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Qi Wang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xinxin Liu
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Feng Wang
- Department of Endoscopy Center, Peking University First Hospital, Beijing, 100034, China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, 100034, China
| |
Collapse
|
25
|
Silvestris N, Argentiero A, Brunetti O, Sonnessa M, Colonna F, Delcuratolo S, Luchini C, Scarpa A, Lonardi S, Nappo F, Fassan M, Solimando AG, Fucci L, Saponaro C. PD-L1 and Notch as novel biomarkers in pancreatic sarcomatoid carcinoma: a pilot study. Expert Opin Ther Targets 2021; 25:1007-1016. [PMID: 34846251 DOI: 10.1080/14728222.2021.2011859] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/24/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The improved immunological understanding revealed the tumor microenvironment as an appealing driver to restore the immune response against cancer cells resulting in a paradigm shift in the oncology field. However, the complexity of the tumor milieu suggests the role of several pathways linking in immunomodulation mechanisms. Pancreatic cancer represents a model of the intricate relationship between malignant cells and their surrounding neighborhood. RESEARCH DESIGN AND METHODS In this study, we analyzed, retrospectively, six cases of rare pancreatic sarcomatoid carcinoma (PSC) and evaluated the expression of PD-L1 and Notch, aiming to explore new attributes in immunophenotype. RESULTS PD-L1 CPS ≥ 1was common in PSCs (83%) with half samples expressing PD-L1 CPS ≥ 50. Notch1 and Notch3 demonstrated a high range of expression. A direct significant correlation between PD-L1 and Notch3 overexpression (r = 0.7; p = 0.036) has been observed. Immunofluorescence studies revealed a co-localization of Notch3 and PD-L1 when both proteins were over-expressed within cytoplasmic or membranous compartments of the same cells. CONCLUSIONS Our data identify a unique biological characterization of this rare pancreatic histotype. These findings provide a rationale for future studies evaluating the potential crosstalk between PD-L1/PD-1 axis and Notch pathways and prompting the development of novel therapeutics strategy.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Antonella Argentiero
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Margherita Sonnessa
- Functional Biomorphology Laboratory, Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Fulvia Colonna
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Sabina Delcuratolo
- Clinical Trial Office IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Claudio Luchini
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Enets Center of Excellence of Verona, Verona, Italy
| | - Aldo Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Enets Center of Excellence of Verona, Verona, Italy
- Arc-net Applied Research on Cancer Centre, University of Verona, Verona, Italy
| | - Sara Lonardi
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology Iov-irccs, Padua, Italy
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology Iov - Irccs, Padua, Italy
| | - Floriana Nappo
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology Iov-irccs, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (Dimed), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
- Veneto Institute of Oncology Iov - Irccs, Padua, Italy
| | - Antonio Giovanni Solimando
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Livia Fucci
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Concetta Saponaro
- Functional Biomorphology Laboratory, Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| |
Collapse
|
26
|
Rahmanuddin S, Korn R, Cridebring D, Borazanci E, Brase J, Boswell W, Jamil A, Cai W, Sabir A, Motarjem P, Koay E, Mitra A, Goel A, Ho J, Chung V, Von Hoff DD. Role of 3D Volumetric and Perfusion Imaging for Detecting Early Changes in Pancreatic Adenocarcinoma. Front Oncol 2021; 11:678617. [PMID: 34568010 PMCID: PMC8456995 DOI: 10.3389/fonc.2021.678617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/13/2021] [Indexed: 12/30/2022] Open
Abstract
Purpose There is a major shortage of reliable early detection methods for pancreatic cancer in high-risk groups. The focus of this preliminary study was to use Time Intensity-Density Curve (TIDC) and Marley Equation analyses, in conjunction with 3D volumetric and perfusion imaging to demonstrate their potential as imaging biomarkers to assist in the early detection of Pancreatic Ductal Adenocarcinoma (PDAC). Experimental Designs A quantitative retrospective and prospective study was done by analyzing multi-phase Computed Tomography (CT) images of 28 patients undergoing treatment at different stages of pancreatic adenocarcinoma using advanced 3D imaging software to identify the perfusion and radio density of tumors. Results TIDC and the Marley Equation proved useful in quantifying tumor aggressiveness. Perfusion delays in the venous phase can be linked to Vascular Endothelial Growth Factor (VEGF)-related activity which represents the active part of the tumor. 3D volume analysis of the multiphase CT scan of the patient showed clear changes in arterial and venous perfusion indicating the aggressive state of the tumor. Conclusion TIDC and 3D volumetric analysis can play a significant role in defining the response of the tumor to treatment and identifying early-stage aggressiveness.
Collapse
Affiliation(s)
- Syed Rahmanuddin
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Ronald Korn
- Virginia G Piper Cancer Center, Honor Health, Scottsdale, AZ, United States
| | - Derek Cridebring
- Molecular Medicine Division, Translational Genomics Research Institute (TGEN), Phoenix, AZ, United States
| | - Erkut Borazanci
- Virginia G Piper Cancer Center, Honor Health, Scottsdale, AZ, United States
| | - Jordyn Brase
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - William Boswell
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Asma Jamil
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Wenli Cai
- Department of Radiology, Massachusetts General Hospital, Boston, MA, United States
| | - Aqsa Sabir
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Pejman Motarjem
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| | - Eugene Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anirban Mitra
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ajay Goel
- Molecular Diagnostic and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, United States
| | - Joyce Ho
- Molecular Diagnostic and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, United States
| | - Vincent Chung
- Molecular Diagnostic and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Monrovia, CA, United States
| | - Daniel D Von Hoff
- National Medical Center & Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, CA, United States.,Virginia G Piper Cancer Center, Honor Health, Scottsdale, AZ, United States.,Molecular Medicine Division, Translational Genomics Research Institute (TGEN), Phoenix, AZ, United States
| |
Collapse
|
27
|
Edwards P, Kang BW, Chau I. Targeting the Stroma in the Management of Pancreatic Cancer. Front Oncol 2021; 11:691185. [PMID: 34336679 PMCID: PMC8316993 DOI: 10.3389/fonc.2021.691185] [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: 04/05/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) presents extremely aggressive tumours and is associated with poor survival. This is attributed to the unique features of the tumour microenvironment (TME), which is known to create a dense stromal formation and poorly immunogenic condition. In particular, the TME of PC, including the stromal cells and extracellular matrix, plays an essential role in the progression and chemoresistance of PC. Consequently, several promising agents that target key components of the stroma have already been developed and are currently in multiple stages of clinical trials. Therefore, the authors review the latest available evidence on novel stroma-targeting approaches, highlighting the potential impact of the stroma as a key component of the TME in PC.
Collapse
Affiliation(s)
- Penelope Edwards
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
28
|
Study of the Mechanism by Which Curcumin Cooperates with Sestrin2 to Inhibit the Growth of Pancreatic Cancer. Gastroenterol Res Pract 2021; 2021:7362233. [PMID: 34306067 PMCID: PMC8266438 DOI: 10.1155/2021/7362233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/15/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Background Pancreatic carcinoma is a malignant tumor with a high fatality rate, and the increased resistance of pancreatic carcinoma to chemotherapy has become a difficult problem in clinical practice. Hence, it is imperative to develop an effective treatment for pancreatic cancer. Sestrins are a class of stress-induced proteins that have antioxidation functions, regulating cell growth and metabolism. Curcumin is a natural pigment isolated from turmeric. Several studies have also suggested that this molecule has multiple pharmacological effects, such as anti-inflammatory, antioxidant, and antitumor effects. However, there are insufficient studies on curcumin cooperating with the sestrin family to inhibit tumors, and the mechanism is still unclear. Our aim was to observe the potential anticancer effects of curcumin combined with the sestrin family on pancreatic carcinoma and probe its possible molecular mechanisms. Methods Lentiviral infection, real-time fluorescence quantitative PCR assays, Cell Counting Kit-8 assays, real-time cell analysis technology, colony formation assays, wound healing assays, Transwell invasion assays, protein extraction, and western blots (WBs) were used to evaluate the effect of curcumin combined with sestrin2 on the proliferation, invasion, and migration of pancreatic carcinoma cells. Results The results revealed that curcumin cooperated with sestrin2 to significantly suppress pancreatic cancer. In addition, we determined that sestrin2 cooperated with curcumin to inhibit pancreatic cancer by specifically targeting Nrf2/Keap1/HO-1/NQO-1. Conclusion These findings clarify that curcumin-mediated synergistic targeting of sestrin2 is a potentially valuable treatment for pancreatic cancer.
Collapse
|
29
|
Antibody therapy in pancreatic cancer: mAb-ye we're onto something? Biochim Biophys Acta Rev Cancer 2021; 1876:188557. [PMID: 33945846 DOI: 10.1016/j.bbcan.2021.188557] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/23/2021] [Accepted: 04/25/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer remains an extremely deadly disease, with little improvement seen in treatment or outcomes over the last 40 years. Targeted monoclonal antibody therapy is one area that has been explored in attempts to tackle this disease. This review examines antibodies that have undergone clinical evaluation in pancreatic cancer. These antibodies target a wide variety of molecules, including tumour cell surface, stromal, immune and embryonic pathway targets. We discuss the therapeutic utility of these therapies both as monotherapeutics and in combination with other treatments such as chemotherapy. While antibody therapy for pancreatic cancer has yet to yield significant success, lessons learned from research thus far highlights future directions that may help overcome observed hurdles to yield clinically efficacious results.
Collapse
|
30
|
Derakhshani A, Rostami Z, Safarpour H, Shadbad MA, Nourbakhsh NS, Argentiero A, Taefehshokr S, Tabrizi NJ, Kooshkaki O, Astamal RV, Singh PK, Taefehshokr N, Alizadeh N, Silvestris N, Baradaran B. From Oncogenic Signaling Pathways to Single-Cell Sequencing of Immune Cells: Changing the Landscape of Cancer Immunotherapy. Molecules 2021; 26:2278. [PMID: 33920054 PMCID: PMC8071039 DOI: 10.3390/molecules26082278] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 12/19/2022] Open
Abstract
Over the past decade, there have been remarkable advances in understanding the signaling pathways involved in cancer development. It is well-established that cancer is caused by the dysregulation of cellular pathways involved in proliferation, cell cycle, apoptosis, cell metabolism, migration, cell polarity, and differentiation. Besides, growing evidence indicates that extracellular matrix signaling, cell surface proteoglycans, and angiogenesis can contribute to cancer development. Given the genetic instability and vast intra-tumoral heterogeneity revealed by the single-cell sequencing of tumoral cells, the current approaches cannot eliminate the mutating cancer cells. Besides, the polyclonal expansion of tumor-infiltrated lymphocytes in response to tumoral neoantigens cannot elicit anti-tumoral immune responses due to the immunosuppressive tumor microenvironment. Nevertheless, the data from the single-cell sequencing of immune cells can provide valuable insights regarding the expression of inhibitory immune checkpoints/related signaling factors in immune cells, which can be used to select immune checkpoint inhibitors and adjust their dosage. Indeed, the integration of the data obtained from the single-cell sequencing of immune cells with immune checkpoint inhibitors can increase the response rate of immune checkpoint inhibitors, decrease the immune-related adverse events, and facilitate tumoral cell elimination. This study aims to review key pathways involved in tumor development and shed light on single-cell sequencing. It also intends to address the shortcomings of immune checkpoint inhibitors, i.e., their varied response rates among cancer patients and increased risk of autoimmunity development, via applying the data from the single-cell sequencing of immune cells.
Collapse
Affiliation(s)
- Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Zeinab Rostami
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Hossein Safarpour
- Cellular & Molecular Research Center, Birjand University of Medical Sciences, Birjand 97178-53577, Iran;
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| | | | | | - Sina Taefehshokr
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Neda Jalili Tabrizi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand 97178-53577, Iran; (Z.R.); (O.K.)
| | - Reza Vaezi Astamal
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Pankaj Kumar Singh
- Principal Research Technologist, Department of Radiation Oncology, Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA;
| | - Nima Taefehshokr
- Department of Microbiology and Immunology, Center for Human Immunology, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
| | - Nicola Silvestris
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 51656-65811, Iran; (A.D.); (M.A.S.); (S.T.); (N.J.T.); (R.V.A.); (N.A.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 51666-14766, Iran
| |
Collapse
|
31
|
Kaur J, Singh P, Enzler T, Sahai V. Emerging antibody therapies for pancreatic adenocarcinoma: a review of recent phase 2 trials. Expert Opin Emerg Drugs 2021; 26:103-129. [PMID: 33734833 DOI: 10.1080/14728214.2021.1905795] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Pancreatic adenocarcinoma is now the third-leading cause of cancer-related deaths in the US which can be attributed to rising incidence, diagnosis at advanced stages and early development of metastasis. Systemic therapy remains palliative with early development of resistance possibly related to the constitutive activation of 'undruggable' KRAS, immunosuppressive microenvironment, and intense desmoplasia. The advancements in molecular biology has led to the development and investigation of targeted and immune therapeutics.Areas covered: This study provides a comprehensive review of the literature to further the understanding of molecular targets with their respective antibody-based therapies in clinical development in pancreatic cancer. PubMed was systematically searched for English-language articles discussing antibody-based therapies under phase 2 clinical trial investigation in pancreatic adenocarcinoma.Expert opinion: PDAC remains highly resistant to chemotherapy with no significant improvement in survival for patients with advanced or metastatic cancer. Unfortunately, the majority of the antibody-based targeted and immune therapeutics have failed to meet their primary efficacy endpoints in early phase trials. However, there are a few promising antibody-based drugs with intriguing preliminary data that merit further investigation, while many more continue to be developed and investigated preclinically, and in early phase trials.
Collapse
Affiliation(s)
- Jasmeet Kaur
- Department of Internal Medicine, Saint Joseph Mercy Oakland Hospital, Pontiac, MI, USA
| | - Paramveer Singh
- Division of Hematology and Oncology, Department of Internal Medicine, Wayne State University, Detroit, MI, USA
| | - Thomas Enzler
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Vaibhav Sahai
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
32
|
Liot S, Balas J, Aubert A, Prigent L, Mercier-Gouy P, Verrier B, Bertolino P, Hennino A, Valcourt U, Lambert E. Stroma Involvement in Pancreatic Ductal Adenocarcinoma: An Overview Focusing on Extracellular Matrix Proteins. Front Immunol 2021; 12:612271. [PMID: 33889150 PMCID: PMC8056076 DOI: 10.3389/fimmu.2021.612271] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide and is predicted to become second in 2030 in industrialized countries if no therapeutic progress is made. Among the different types of pancreatic cancers, Pancreatic Ductal Adenocarcinoma (PDAC) is by far the most represented one with an occurrence of more than 90%. This specific cancer is a devastating malignancy with an extremely poor prognosis, as shown by the 5-years survival rate of 2–9%, ranking firmly last amongst all cancer sites in terms of prognostic outcomes for patients. Pancreatic tumors progress with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. This malignancy is characterized by an extremely dense stroma deposition around lesions, accompanied by tissue hypovascularization and a profound immune suppression. Altogether, these combined features make access to cancer cells almost impossible for conventional chemotherapeutics and new immunotherapeutic agents, thus contributing to the fatal outcomes of the disease. Initially ignored, the Tumor MicroEnvironment (TME) is now the subject of intensive research related to PDAC treatment and could contain new therapeutic targets. In this review, we will summarize the current state of knowledge in the field by focusing on TME composition to understand how this specific compartment could influence tumor progression and resistance to therapies. Attention will be paid to Tenascin-C, a matrix glycoprotein commonly upregulated during cancer that participates to PDAC progression and thus contributes to poor prognosis.
Collapse
Affiliation(s)
- Sophie Liot
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Jonathan Balas
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Alexandre Aubert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Laura Prigent
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Perrine Mercier-Gouy
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Philippe Bertolino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ana Hennino
- Cancer Research Center of Lyon, UMR INSERM 1052, CNRS 5286, Lyon, France
| | - Ulrich Valcourt
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Elise Lambert
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR CNRS 5305, Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| |
Collapse
|
33
|
Ahangar NK, Hemmat N, Khalaj-Kondori M, Shadbad MA, Sabaie H, Mokhtarzadeh A, Alizadeh N, Derakhshani A, Baghbanzadeh A, Dolatkhah K, Silvestris N, Baradaran B. The Regulatory Cross-Talk between microRNAs and Novel Members of the B7 Family in Human Diseases: A Scoping Review. Int J Mol Sci 2021; 22:2652. [PMID: 33800752 PMCID: PMC7962059 DOI: 10.3390/ijms22052652] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/21/2021] [Accepted: 03/02/2021] [Indexed: 12/18/2022] Open
Abstract
The members of the B7 family, as immune checkpoint molecules, can substantially regulate immune responses. Since microRNAs (miRs) can regulate gene expression post-transcriptionally, we conducted a scoping review to summarize and discuss the regulatory cross-talk between miRs and new B7 family immune checkpoint molecules, i.e., B7-H3, B7-H4, B7-H5, butyrophilin like 2 (BTNL2), B7-H6, B7-H7, and immunoglobulin like domain containing receptor 2 (ILDR2). The current study was performed using a six-stage methodology structure and Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. PubMed, Embase, Scopus, Cochrane, ProQuest, and Google Scholar were systematically searched to obtain the relevant records to 5 November 2020. Two authors independently reviewed the obtained records and extracted the desired data. After quantitative and qualitative analyses, we used bioinformatics approaches to extend our knowledge about the regulatory cross-talk between miRs and the abovementioned B7 family members. Twenty-seven articles were identified that fulfilled the inclusion criteria. Studies with different designs reported gene-miR regulatory axes in various cancer and non-cancer diseases. The regulatory cross-talk between the aforementioned B7 family molecules and miRs might provide valuable insights into the pathogenesis of various human diseases.
Collapse
Affiliation(s)
- Noora Karim Ahangar
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 5166616471, Iran; (N.K.A.); (M.K.-K.)
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 5166616471, Iran; (N.K.A.); (M.K.-K.)
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Hani Sabaie
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
- IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Katayoun Dolatkhah
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| | - Nicola Silvestris
- IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (N.H.); (M.A.S.); (A.M.); (N.A.); (A.D.); (A.B.); (K.D.)
| |
Collapse
|
34
|
Chen K, Wang Q, Kornmann M, Tian X, Yang Y. The Role of Exosomes in Pancreatic Cancer From Bench to Clinical Application: An Updated Review. Front Oncol 2021; 11:644358. [PMID: 33718244 PMCID: PMC7952979 DOI: 10.3389/fonc.2021.644358] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most dismal gastrointestinal malignancies with an overall 5-year survival rate of 8%-9%. The intra-tumor heterogeneity and special tumor microenvironment in PDAC make it challenging to develop effective treatment strategies. Exosomes are extracellular vesicles that originate from the endosomes and have a diameter of 40-160 nm. A growing body of evidence has shown that exosomes play vital roles in tumor initiation and development. Recently, extensive application of exosomes as biomarkers and drug carriers has rendered them attractive in the field of PDAC. This review summarizes the latest progress in the methodologies for isolation, modification, and tracking of exosomes, exosome-mediated cell-to-cell communication, clinical applications of exosome as minimally invasive liquid biopsy and drugs carriers, as well as their involvement in the angiogenic regulation in PDAC. In spite of these advancements, some obstacles are still required to be overcome to use the exosome-based technologies for early diagnosis or improvement of prognosis of patients with PDAC.
Collapse
Affiliation(s)
- Kai Chen
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Qi Wang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Marko Kornmann
- Clinic of General, Visceral and Transplantation Surgery, University of Ulm, Ulm, Germany
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yinmo Yang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
35
|
Yamakawa K, Ye J, Nakano-Narusawa Y, Matsuda Y. Pathological Changes in Pancreatic Carcinogenesis: A Review. Cancers (Basel) 2021; 13:cancers13040686. [PMID: 33567676 PMCID: PMC7914468 DOI: 10.3390/cancers13040686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Despite advances in diagnostics and therapeutics, the prognosis of pancreatic cancer remains dismal. Because of a lack of early diagnostic methods, aggressive local progression, and high incidence of distant metastasis, most pancreatic cancers are inoperable; therefore, the characteristics of early pancreatic cancer have not been well understood. Autopsy studies revealed the characteristics of prediagnostic pancreatic malignancies, including precancerous lesions, early stage pancreatic cancer, and pancreatic cancer without clinical symptoms (occult cancers). Animal models using hamsters and genetically engineered mice have focused on mechanisms of carcinogenesis, thereby providing insights into risk factors and prevention and serving as a preclinical test for the development of novel diagnostic and treatment modalities. In this review, we have summarized pathological changes in the pancreas of humans and experimental animals during carcinogenesis.
Collapse
Affiliation(s)
| | | | | | - Yoko Matsuda
- Correspondence: ; Tel.: +81-87-891-2109; Fax: +81-87-891-2112
| |
Collapse
|
36
|
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: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 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
|
37
|
Suri R, Zimmerman JW, Burkhart RA. Modeling human pancreatic ductal adenocarcinoma for translational research: current options, challenges, and prospective directions. ANNALS OF PANCREATIC CANCER 2020; 3:17. [PMID: 33889840 PMCID: PMC8059695 DOI: 10.21037/apc-20-29] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with one of the lowest survival rates. Early detection, an improved understanding of tumor biology, and novel therapeutic discoveries are needed in order to improve overall patient survival. Scientific progress towards meeting these goals relies upon accurate modeling of the human disease. From two-dimensional (2D) cell lines to the advanced modeling available today, we aim to characterize the critical tools in efforts to further understand PDAC biology. The National Center for Biotechnology Information's PubMed and the Elsevier's SCOPUS were used to perform a comprehensive literature review evaluating preclinical human-derived PDAC models. Keywords included pancreatic cancer, PDAC, preclinical models, KRAS mutations, xenograft, co-culturing fibroblasts, co-culturing lymphocytes and PDAC immunotherapy Initial search was limited to articles about PDAC and was then expanded to include other gastrointestinal malignancies where information may complement our effort. A supervised review of the key literature's references was utilized to augment the capture of relevant data. The discovery and refinement of techniques enabling immortalized 2D cell culture provided the cornerstone for modern cancer biology research. Cell lines have been widely used to represent PDAC in vitro but are limited in capacity to model three-dimensional (3D) tumor attributes and interactions within the tumor microenvironment. Xenografts are an alternative method to model PDAC with improved capacity to understand certain aspects of 3D tumor biology in vivo while limited by the use of immunodeficient mice. Advances of in vitro modeling techniques have led to 3D organoid models for PDAC biology. Co-culturing models in the 3D environment have been proposed as an efficient modeling system for improving upon the limitations encountered in the standard 2D and xenograft tumor models. The integrated network of cells and stroma that comprise PDAC in vivo need to be accurately depicted ex vivo to continue to make progress in this disease. Recapitulating the complex tumor microenvironment in a preclinical model of human disease is an outstanding and urgent need in PDAC. Definitive characterization of available human models for PDAC serves to further the core mission of pancreatic cancer translational research.
Collapse
Affiliation(s)
- Reecha Suri
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Department of Medical Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| | - Richard A. Burkhart
- Division of Hepatobiliary and Pancreatic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
38
|
Targeting TGF-β-Mediated SMAD Signaling Pathway via Novel Recombinant Cytotoxin II: A Potent Protein from Naja naja oxiana Venom in Melanoma. Molecules 2020; 25:molecules25215148. [PMID: 33167431 PMCID: PMC7663949 DOI: 10.3390/molecules25215148] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/31/2022] Open
Abstract
Since the current treatments have not resulted in the desired outcomes for melanoma patients, there is a need to identify more effective medications. Together with other snake venom proteins, cytotoxin-II has shown promising results in tumoral cells. In this study, recombinant cytotoxin-II (rCTII) was expressed in SHuffle® T7 Express cells, while the epitope mapping of rCTII was performed to reveal the antibody-binding regions of rCTII. The MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay was used to assess the viability of SK-MEL-3 and HFF-2 cells after treating these cells with rCTII. The qRT-PCR was performed to evaluate the expression levels of matrix metallopeptidase 3 (MMP-3), SMAD2, SMAD3, caspase-8, caspase-9, and miR-214 in order to reveal the rCTII-induced signaling pathways in melanoma. Our results have shown that two regions of amino acids, 6-16 and 19-44, as predicted epitopes of this toxin, are essential for understanding the toxicity of rCTII. Treating the melanoma cells with rCTII substantially inhibited the transforming growth factor-beta (TGF-β)-SMAD signaling pathway and down-regulated the expression of MMP-3 and miR-214 as well. This cytotoxin also restored apoptosis mainly via the intrinsic pathway. The down-regulation of MMP-3 and miR-214 might be associated with the anti-metastatic property of rCTII in melanoma. The inhibitory effect of rCTII on the TGF-β signaling pathway might be associated with increased apoptosis and decreased cancer cell proliferation. It is interesting to see that the IC50 value of rCTII has been lower in the melanoma cells than non-tumoral cells, which may indicate its potential effects as a drug. In conclusion, rCTII, as a novel medication, might serve as a potent and efficient anticancer drug in melanoma.
Collapse
|
39
|
Immune Checkpoints and CAR-T Cells: The Pioneers in Future Cancer Therapies? Int J Mol Sci 2020; 21:ijms21218305. [PMID: 33167514 PMCID: PMC7663909 DOI: 10.3390/ijms21218305] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
Although the ever-increasing number of cancer patients pose substantial challenges worldwide, finding a treatment with the highest response rate and the lowest number of side effects is still undergoing research. Compared to chemotherapy, the relatively low side effects of cancer immunotherapy have provided ample opportunity for immunotherapy to become a promising approach for patients with malignancy. However, the clinical translation of immune-based therapies requires robust anti-tumoral immune responses. Immune checkpoints have substantial roles in the induction of an immunosuppressive tumor microenvironment and tolerance against tumor antigens. Identifying and targeting these inhibitory axes, which can be established between tumor cells and tumor-infiltrating lymphocytes, can facilitate the development of anti-tumoral immune responses. Bispecific T-cell engagers, which can attract lymphocytes to the tumor microenvironment, have also paved the road for immunological-based tumor elimination. The development of CAR-T cells and their gene editing have brought ample opportunity to recognize tumor antigens, independent from immune checkpoints and the major histocompatibility complex (MHC). Indeed, there have been remarkable advances in developing various CAR-T cells to target tumoral cells. Knockout of immune checkpoints via gene editing in CAR-T cells might be designated for a breakthrough for patients with malignancy. In the midst of this fast progress in cancer immunotherapies, there is a need to provide up-to-date information regarding immune checkpoints, bispecific T-cell engagers, and CAR-T cells. Therefore, this review aims to provide recent findings of immune checkpoints, bispecific T-cell engagers, and CAR-T cells in cancer immunotherapy and discuss the pertained clinical trials.
Collapse
|
40
|
Abstract
Adenosquamous carcinoma (ASC), containing both adenocarcinoma and squamous cell carcinoma components, is rare in the digestive system. Limited data is available on ASC of the digestive system (AS-ASC), and the current evidence is available mainly in the form of case reports and case series. We performed a thorough search of the available literature and compiled a review on the epidemiology, histopathology, pathogenesis, clinical manifestations, diagnosis, treatment, and prognosis of AS-ASC. Non-specific clinical and imaging presentations and low diagnostic accuracy of biopsy lead to difficulties in preoperative diagnosis in a high proportion of patients and high malignancy. The pathogenesis remains obscure. Surgery remains the mainstay of treatment for AS-ASC. The role of chemoradiotherapy as an adjuvant treatment is still inconclusive. Key messages Metastatic linings and the lack of efficacious treatments lead to an unfavorable outcome in AS-ASC patients. Further research could help us understand the pathophysiology of AS-ASCand the unique needs of AS-ASC patients.
Collapse
Affiliation(s)
- Hong-Shuai Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Tao He
- Department of Radiation Oncology, Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Li-Li Yang
- Department of Medical Oncology, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
41
|
Randazzo O, Papini F, Mantini G, Gregori A, Parrino B, Liu DSK, Cascioferro S, Carbone D, Peters GJ, Frampton AE, Garajova I, Giovannetti E. "Open Sesame?": Biomarker Status of the Human Equilibrative Nucleoside Transporter-1 and Molecular Mechanisms Influencing its Expression and Activity in the Uptake and Cytotoxicity of Gemcitabine in Pancreatic Cancer. Cancers (Basel) 2020; 12:3206. [PMID: 33142664 PMCID: PMC7692081 DOI: 10.3390/cancers12113206] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 01/14/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive tumor characterized by early invasiveness, rapid progression and resistance to treatment. For more than twenty years, gemcitabine has been the main therapy for PDAC both in the palliative and adjuvant setting. After the introduction of FOLFIRINOX as an upfront treatment for metastatic disease, gemcitabine is still commonly used in combination with nab-paclitaxel as an alternative first-line regimen, as well as a monotherapy in elderly patients unfit for combination chemotherapy. As a hydrophilic nucleoside analogue, gemcitabine requires nucleoside transporters to permeate the plasma membrane, and a major role in the uptake of this drug is played by human equilibrative nucleoside transporter 1 (hENT-1). Several studies have proposed hENT-1 as a biomarker for gemcitabine efficacy in PDAC. A recent comprehensive multimodal analysis of hENT-1 status evaluated its predictive role by both immunohistochemistry (with five different antibodies), and quantitative-PCR, supporting the use of the 10D7G2 antibody. High hENT-1 levels observed with this antibody were associated with prolonged disease-free status and overall-survival in patients receiving gemcitabine adjuvant chemotherapy. This commentary aims to critically discuss this analysis and lists molecular factors influencing hENT-1 expression. Improved knowledge on these factors should help the identification of subgroups of patients who may benefit from specific therapies and overcome the limitations of traditional biomarker studies.
Collapse
Affiliation(s)
- Ornella Randazzo
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, 90123 Palermo, Italy; (B.P.); (S.C.); (D.C.)
| | - Filippo Papini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
| | - Giulia Mantini
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| | - Alessandro Gregori
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, 90123 Palermo, Italy; (B.P.); (S.C.); (D.C.)
| | - Daniel S. K. Liu
- Division of Cancer, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital campus, London W12 0NN, UK;
| | - Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, 90123 Palermo, Italy; (B.P.); (S.C.); (D.C.)
| | - Daniela Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, 90123 Palermo, Italy; (B.P.); (S.C.); (D.C.)
| | - Godefridus J. Peters
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Adam E. Frampton
- Division of Cancer, Department of Surgery & Cancer, Imperial College, Hammersmith Hospital campus, London W12 0NN, UK;
- Faculty of Health and Medical Sciences, The Leggett Building, University of Surrey, Guildford GU2 7XH, UK
| | - Ingrid Garajova
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
- Medical Oncology Unit, University Hospital of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc), 1081 HV Amsterdam, The Netherlands; (O.R.); (F.P.); (G.M.); (A.G.); (G.J.P.); (I.G.)
- Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza, 56017 Pisa, Italy
| |
Collapse
|
42
|
Benjanuwattra J, Chaiyawat P, Pruksakorn D, Koonrungsesomboon N. Therapeutic potential and molecular mechanisms of mycophenolic acid as an anticancer agent. Eur J Pharmacol 2020; 887:173580. [PMID: 32949604 DOI: 10.1016/j.ejphar.2020.173580] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 12/14/2022]
Abstract
Mycophenolic acid (MPA) is the active metabolite of mycophenolate mofetil (MMF), an immunosuppressive drug approved for the prophylaxis of allograft rejection in transplant recipients. Recent advances in the role of the type II isoform of inosine-5'-monophosphate dehydrogenase (IMPDH2) in the tumorigenesis of various types of cancer have called for a second look of MPA, the first IMPDH2 inhibitor discovered a hundred years ago, to be repurposed as an anticancer agent. Over a half century, a number of in vitro and in vivo experiments have consistently shown anticancer activity of MPA against several cell lines obtained from different malignancies and murine models. However, a few clinical trials have been conducted to investigate its anticancer activity in humans, and most of which have shown unsatisfactory results. Understanding of available evidence and underlying mechanism of action is a key step to be done so as to facilitate further investigations of MPA to reach its full therapeutic potential as an anticancer agent. This article provides a comprehensive review of non-clinical and clinical evidence available to date, with the emphasis on the molecular mechanism of action in which MPA exerts its anticancer activities: induction of apoptosis, induction of cell cycle arrest, and alteration of tumor microenvironment. Future perspective for further development of MPA to be an anticancer agent is extensively discussed, with the aim of translating the anticancer property of MPA from bench to bedside.
Collapse
Affiliation(s)
| | - Parunya Chaiyawat
- Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand; Omics Center for Health Sciences (OCHS), Faculty of Medicine, Chiang Mai University, Thailand
| | - Dumnoensun Pruksakorn
- Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand; Omics Center for Health Sciences (OCHS), Faculty of Medicine, Chiang Mai University, Thailand; Biomedical Engineering Institute, Chiang Mai University, Thailand
| | - Nut Koonrungsesomboon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Thailand; Musculoskeletal Science and Translational Research Center (MSTR), Faculty of Medicine, Chiang Mai University, Thailand.
| |
Collapse
|
43
|
Arepally A, Chomas J, Katz SC, Jaroch D, Kolli KP, Prince E, Liddell RP. Pressure-Enabled Drug Delivery Approach in the Pancreas with Retrograde Venous Infusion of Lipiodol with Ex Vivo Analysis. Cardiovasc Intervent Radiol 2020; 44:141-149. [PMID: 32895782 PMCID: PMC7728652 DOI: 10.1007/s00270-020-02625-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 08/07/2020] [Indexed: 01/03/2023]
Abstract
Purpose To determine the safety and feasibility of pancreatic retrograde venous infusion (PRVI) utilizing a microvalvular infusion system (MVI) to deliver ethiodized oil (lipiodol) by means of the Pressure-Enabled Drug Delivery (PEDD) approach. Methods Utilizing transhepatic access, mapping of the pancreatic body and head venous anatomy was performed in 10 swine. PEDD was performed by cannulation of veins in the head (n = 4) and body (n = 10) of the pancreas with a MVI (Surefire® Infusion System (SIS), Surefire Medical, Inc (DBA TriSalus™ Life Sciences)) followed by infusion with lipiodol. Sets of animals were killed either immediately (n = 8) or at 4 days post-PRVI (n = 2). All pancreata were harvested and studied with micro-CT and histology. We also performed three-dimensional volumetric/multiplanar imaging to assess the vascular distribution of lipiodol within the glands. Results A total of 14 pancreatic veins were successfully infused with an average of 1.7 (0.5–2.0) mL of lipiodol. No notable change in serum chemistries was seen at 4 days. The signal-to-noise ratio (SNR) of lipiodol deposition was statistically increased both within the organ in target relative to non-target pancreatic tissue and compared to extra pancreatic tissue (p < 0.05). Histological evaluation demonstrated no evidence of pancreatic edema or ischemia. Conclusions PEDD using the RVI approach for targeted pancreatic infusions is technically feasible and did not result in organ damage in this pilot animal study.
Collapse
Affiliation(s)
- Aravind Arepally
- Division of Interventional Radiology, Piedmont Radiology, Piedmont Healthcare, 1984 Peachtree Road, Suite 505, Atlanta, Georgia, 30309, USA.
| | - James Chomas
- Formerly TriSalus Life Sciences, Inc, Westminster, USA
| | - Steven C Katz
- Office of Therapeutic Development and Department of Surgery, Roger Williams Medical Center, Providence, USA.,Department of Surgery, Boston University School of Medicine, Boston, USA
| | | | - K Pallav Kolli
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, USA
| | - Ethan Prince
- Radiology, Roger Williams Medical Center, Providence, USA
| | - Robert P Liddell
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, USA
| |
Collapse
|
44
|
Tawila AM, Sun S, Kim MJ, Omar AM, Dibwe DF, Ueda JY, Toyooka N, Awale S. Chemical constituents of Callistemon citrinus from Egypt and their antiausterity activity against PANC-1 human pancreatic cancer cell line. Bioorg Med Chem Lett 2020; 30:127352. [PMID: 32631550 DOI: 10.1016/j.bmcl.2020.127352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/25/2020] [Accepted: 06/12/2020] [Indexed: 01/03/2023]
Abstract
Human pancreatic cancer is resistant to almost all conventional chemotherapeutic agents. It is known to proliferate aggressively within hypovascular tumor microenvironment by exhibiting remarkable tolerance to nutrition starvation, a phenomenon termed as "austerity". Search for the new agents that eliminate the tolerance of cancer cells to nutrition starvation is a promising strategy in anticancer drug discovery. In this study, two new meroterpenoids named callistrilones O and P (1 and 2) together with eight known triterpenes (3-10) were isolated from the active dichloromethane extract of Callistemon citrinus leaves. The structure elucidation of the new compounds was achieved by HRFABMS, 1D, 2D NMR, and ECD quantum calculations. All isolated compounds were tested for their preferential cytotoxicity against PANC-1 human pancreatic cancer cells. Among these, callistrilone O (1) exhibited the most potent preferential cytotoxicity with a PC50 value of 0.3 nM, the strongest activity with over 2000 times potent than the positive control arctigenin. Callistrilone O (1) induced dramatic alterations in PANC-1 cell morphology leading to cell death under nutrient-deprived conditions. Compound 1 also inhibited PANC-1 cell migration and -PANC-1 colony formation under the nutrient-rich condition.
Collapse
Affiliation(s)
- Ahmed M Tawila
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Sijia Sun
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Min Jo Kim
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Ashraf M Omar
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Dya Fita Dibwe
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Jun-Ya Ueda
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1, Hirokoshingai, Kure, Hiroshima 737-0112, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, Department of Bio-functional Molecular Engineering, University of Toyama, Toyama 930-8555, Japan; Faculty of Engineering, University of Toyama, Toyama 930-8555, Japan
| | - Suresh Awale
- Division of Natural Drug Discovery, Institute of Natural Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
45
|
Enhancing the Efficacy of CAR T Cells in the Tumor Microenvironment of Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12061389. [PMID: 32481570 PMCID: PMC7353070 DOI: 10.3390/cancers12061389] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/23/2020] [Accepted: 05/26/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer has the worst prognosis and lowest survival rate among all types of cancers and thus, there exists a strong need for novel therapeutic strategies. Chimeric antigen receptor (CAR)-modified T cells present a new potential option after successful FDA-approval in hematologic malignancies, however, current CAR T cell clinical trials in pancreatic cancer failed to improve survival and were unable to demonstrate any significant response. The physical and environmental barriers created by the distinct tumor microenvironment (TME) as a result of the desmoplastic reaction in pancreatic cancer present major hurdles for CAR T cells as a viable therapeutic option in this tumor entity. Cancer cells and cancer-associated fibroblasts express extracellular matrix molecules, enzymes, and growth factors, which can attenuate CAR T cell infiltration and efficacy. Recent efforts demonstrate a niche shift where targeting the TME along CAR T cell therapy is believed or hoped to provide a substantial clinical added value to improve overall survival. This review summarizes therapeutic approaches targeting the TME and their effect on CAR T cells as well as their outcome in preclinical and clinical trials in pancreatic cancer.
Collapse
|
46
|
Gupta P, Pérez-Mancera PA, Kocher H, Nisbet A, Schettino G, Velliou EG. A Novel Scaffold-Based Hybrid Multicellular Model for Pancreatic Ductal Adenocarcinoma-Toward a Better Mimicry of the in vivo Tumor Microenvironment. Front Bioeng Biotechnol 2020; 8:290. [PMID: 32391339 PMCID: PMC7193232 DOI: 10.3389/fbioe.2020.00290] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/19/2020] [Indexed: 12/11/2022] Open
Abstract
With a very low survival rate, pancreatic ductal adenocarcinoma (PDAC) is a deadly disease. This has been primarily attributed to (i) its late diagnosis and (ii) its high resistance to current treatment methods. The latter specifically requires the development of robust, realistic in vitro models of PDAC, capable of accurately mimicking the in vivo tumor niche. Advancements in the field of tissue engineering (TE) have helped the development of such models for PDAC. Herein, we report for the first time a novel hybrid, polyurethane (PU) scaffold-based, long-term, multicellular (tri-culture) model of pancreatic cancer involving cancer cells, endothelial cells, and stellate cells. Recognizing the importance of ECM proteins for optimal growth of different cell types, the model consists of two different zones/compartments: an inner tumor compartment consisting of cancer cells [fibronectin (FN)-coated] and a surrounding stromal compartment consisting of stellate and endothelial cells [collagen I (COL)-coated]. Our developed novel hybrid, tri-culture model supports the proliferation of all different cell types for 35 days (5 weeks), which is the longest reported timeframe in vitro. Furthermore, the hybrid model showed extensive COL production by the cells, mimicking desmoplasia, one of PDAC's hallmark features. Fibril alignment of the stellate cells was observed, which attested to their activated state. All three cell types expressed various cell-specific markers within the scaffolds, throughout the culture period and showed cellular migration between the two zones of the hybrid scaffold. Our novel model has great potential as a low-cost tool for in vitro studies of PDAC, as well as for treatment screening.
Collapse
Affiliation(s)
- Priyanka Gupta
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, United Kingdom
| | - Pedro A. Pérez-Mancera
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Hemant Kocher
- Centre for Tumour Biology and Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Andrew Nisbet
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Giuseppe Schettino
- Department of Physics, University of Surrey, Guildford, United Kingdom
- Medical Radiation Science Group, The National Physical Laboratory, Teddington, United Kingdom
| | - Eirini G. Velliou
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford, United Kingdom
| |
Collapse
|
47
|
Momeny M, Alishahi Z, Eyvani H, Esmaeili F, Zaghal A, Ghaffari P, Tavakkoly-Bazzaz J, Alimoghaddam K, Ghavamzadeh A, Ghaffari SH. Anti-tumor activity of cediranib, a pan-vascular endothelial growth factor receptor inhibitor, in pancreatic ductal adenocarcinoma cells. Cell Oncol (Dordr) 2020; 43:81-93. [DOI: 10.1007/s13402-019-00473-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2019] [Indexed: 12/18/2022] Open
|
48
|
Pezzilli R, Caccialanza R, Capurso G, Brunetti O, Milella M, Falconi M. Pancreatic Enzyme Replacement Therapy in Pancreatic Cancer. Cancers (Basel) 2020; 12:275. [PMID: 31979186 PMCID: PMC7073203 DOI: 10.3390/cancers12020275] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/19/2022] Open
Abstract
Pancreatic cancer is an aggressive malignancy and the seventh leading cause of global cancer deaths in industrialised countries. More than 80% of patients suffer from significant weight loss at diagnosis and over time tend to develop severe cachexia. A major cause of weight loss is malnutrition. Patients may experience pancreatic exocrine insufficiency (PEI) before diagnosis, during nonsurgical treatment, and/or following surgery. PEI is difficult to diagnose because testing is cumbersome. Consequently, PEI is often detected clinically, especially in non-specialised centres, and treated empirically. In this position paper, we review the current literature on nutritional support and pancreatic enzyme replacement therapy (PERT) in patients with operable and non-operable pancreatic cancer. To increase awareness on the importance of PERT in pancreatic patients, we provide recommendations based on literature evidence, and when data were lacking, based on our own clinical experience.
Collapse
Affiliation(s)
- Raffaele Pezzilli
- Gastroenterology Unit, San Carlo Hospital, Via P. Petrone, 85100 Potenza, Italy
| | - Riccardo Caccialanza
- Clinical Nutrition and Dietetics Unit, Fondazione IRCCS Policlinico San Matteo, Viale Camillo Golgi 19, 27100 Pavia, Italy;
| | - Gabriele Capurso
- Clinical Research, Pancreato-Biliary Endoscopy and EUS Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy;
| | - Oronzo Brunetti
- Medical Oncology Unit, National Cancer Institute “Giovanni Paolo II”, Viale O. Flacco 65, 70124 Bari, Italy;
| | - Michele Milella
- Residency Program in Medical Oncology, University of Verona, Via S. Francesco 22, 37129 Verona, Italy;
- AOUI Verona, Sede Policlinico Universitario G.B. Rossi Borgo Roma, P.le L.A. Scuro 10, 37134 Verona, Italy
| | - Massimo Falconi
- Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milano, Italy;
| |
Collapse
|
49
|
Dosch AR, Dai X, Reyzer ML, Mehra S, Srinivasan S, Willobee BA, Kwon D, Kashikar N, Caprioli R, Merchant NB, Nagathihalli NS. Combined Src/EGFR Inhibition Targets STAT3 Signaling and Induces Stromal Remodeling to Improve Survival in Pancreatic Cancer. Mol Cancer Res 2020; 18:623-631. [PMID: 31949002 DOI: 10.1158/1541-7786.mcr-19-0741] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 11/26/2019] [Accepted: 01/10/2020] [Indexed: 02/07/2023]
Abstract
Lack of durable response to cytotoxic chemotherapy is a major contributor to the dismal outcomes seen in pancreatic ductal adenocarcinoma (PDAC). Extensive tumor desmoplasia and poor vascular supply are two predominant characteristics which hinder the delivery of chemotherapeutic drugs into PDAC tumors and mediate resistance to therapy. Previously, we have shown that STAT3 is a key biomarker of therapeutic resistance to gemcitabine treatment in PDAC, which can be overcome by combined inhibition of the Src and EGFR pathways. Although it is well-established that concurrent EGFR and Src inhibition exert these antineoplastic properties through direct inhibition of mitogenic pathways in tumor cells, the influence of this combined therapy on stromal constituents in PDAC tumors remains unknown. In this study, we demonstrate in both orthotopic tumor xenograft and Ptf1acre/+;LSL-KrasG12D/+;Tgfbr2flox/flox (PKT) mouse models that concurrent EGFR and Src inhibition abrogates STAT3 activation, increases microvessel density, and prevents tissue fibrosis in vivo. Furthermore, the stromal changes induced by parallel EGFR and Src pathway inhibition resulted in improved overall survival in PKT mice when combined with gemcitabine. As a phase I clinical trial utilizing concurrent EGFR and Src inhibition with gemcitabine has recently concluded, these data provide timely translational insight into the novel mechanism of action of this regimen and expand our understanding into the phenomenon of stromal-mediated therapeutic resistance. IMPLICATIONS: These findings demonstrate that Src/EGFR inhibition targets STAT3, remodels the tumor stroma, and results in enhanced delivery of gemcitabine to improve overall survival in a mouse model of PDAC.
Collapse
Affiliation(s)
- Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Xizi Dai
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Michelle L Reyzer
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Siddharth Mehra
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Supriya Srinivasan
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Brent A Willobee
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Deukwoo Kwon
- Department of Public Health, University of Miami Miller School of Medicine, Miami, Florida
| | - Nilesh Kashikar
- Department of Pathology, University of Colorado, Denver, Colorado
| | - Richard Caprioli
- Mass Spectrometry Research Center, Vanderbilt University, Nashville, Tennessee
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida. .,Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| |
Collapse
|
50
|
Ge Y, Westphalen CB, Ma WW, Vega KJ, Weygant N. Implications for Tumor Microenvironment and Epithelial Crosstalk in the Management of Gastrointestinal Cancers. JOURNAL OF ONCOLOGY 2019; 2019:4835318. [PMID: 32082375 PMCID: PMC7012231 DOI: 10.1155/2019/4835318] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/23/2019] [Accepted: 10/12/2019] [Indexed: 02/08/2023]
Abstract
Rapid advances in technology are revealing previously unknown organization, cooperation, and limitations within the population of nontumor cells surrounding the tumor epithelium known as the tumor microenvironment (TME). Nowhere are these findings more pertinent than in the gastrointestinal (GI) tract where exquisite cell specialization supports a complex microenvironmental niche characterized by rapid stemness-associated cell turnover, pathogen sensing, epithelial orchestration of immune signaling, and other facets that maintain the complex balance between homeostasis, inflammation, and disease. Here, we summarize and discuss select emerging concepts in the precancerous microenvironment, TME, and tumor epithelial-TME crosstalk as well as their implications for the management of GI cancers.
Collapse
Affiliation(s)
- Yang Ge
- Dept of Oncology, Beijing Chao-Yang Hospital, Capital Medical Univ., Beijing, China
| | | | - Wen Wee Ma
- Dept of Oncology, Mayo Clinic, Minneapolis, MN, USA
| | - Kenneth J. Vega
- Dept of Gastroenterology, Augusta University, Augusta, GA, USA
| | - Nathaniel Weygant
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|