1
|
Avsharian LC, Loganathan S, Ebelt ND, Shalamzari AF, Rodarte Muñoz I, Manuel ER. Tumor-Colonizing E. coli Expressing Both Collagenase and Hyaluronidase Enhances Therapeutic Efficacy of Gemcitabine in Pancreatic Cancer Models. Biomolecules 2024; 14:1458. [PMID: 39595636 PMCID: PMC11591662 DOI: 10.3390/biom14111458] [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: 10/28/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Desmoplasia is a hallmark feature of pancreatic ductal adenocarcinoma (PDAC) that contributes significantly to treatment resistance. Approaches to enhance drug delivery into fibrotic PDAC tumors continue to be an important unmet need. In this study, we have engineered a tumor-colonizing E. coli-based agent that expresses both collagenase and hyaluronidase as a strategy to reduce desmoplasia and enhance the intratumoral perfusion of anticancer agents. Overall, we observed that the tandem expression of both these enzymes by tumor-colonizing E. coli resulted in the reduced presence of intratumoral collagen and hyaluronan, which likely contributed to the enhanced chemotherapeutic efficacy observed when used in combination. These results highlight the importance of combination treatments involving the depletion of desmoplastic components in PDAC before or during treatment.
Collapse
Affiliation(s)
- Lara C. Avsharian
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA;
| | - Suvithanandhini Loganathan
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Nancy D. Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Azadeh F. Shalamzari
- Irell and Manella Graduate School of Biological Sciences, City of Hope, Duarte, CA 91010, USA;
| | - Itzel Rodarte Muñoz
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| | - Edwin R. Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, CA 91010, USA; (L.C.A.); (S.L.); (N.D.E.)
| |
Collapse
|
2
|
Doctor A, Laube M, Meister S, Kiss OC, Kopka K, Hauser S, Pietzsch J. Combined PET Radiotracer Approach Reveals Insights into Stromal Cell-Induced Metabolic Changes in Pancreatic Cancer In Vitro and In Vivo. Cancers (Basel) 2024; 16:3393. [PMID: 39410013 PMCID: PMC11475921 DOI: 10.3390/cancers16193393] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Background/Objective Pancreatic stellate cells (PSCs) in pancreatic adenocarcinoma (PDAC) are producing extracellular matrix, which promotes the formation of a dense fibrotic microenvironment. This makes PDAC a highly heterogeneous tumor-stroma-driven entity, associated with reduced perfusion, limited oxygen supply, high interstitial fluid pressure, and limited bioavailability of therapeutic agents. Methods In this study, spheroid and tumor xenograft models of human PSCs and PanC-1 cells were characterized radiopharmacologically using a combined positron emission tomography (PET) radiotracer approach. [18F]FDG, [18F]FMISO, and [18F]FAPI-74 were employed to monitor metabolic activity, hypoxic metabolic state, and functional expression of fibroblast activation protein alpha (FAPα), a marker of activated PSCs. Results In vitro, PanC-1 and multi-cellular tumor spheroids demonstrated comparable glucose uptake and hypoxia, whereas FAPα expression was significantly higher in PSC spheroids. In vivo, glucose uptake as well as the transition to hypoxia were comparable in PanC-1 and multi-cellular xenograft models. In mice injected with PSCs, FAPα expression decreased over a period of four weeks post-injection, which was attributed to the successive death of PSCs. In contrast, FAPα expression increased in both PanC-1 and multi-cellular xenograft models over time due to invasion of mouse fibroblasts. Conclusion The presented models are suitable for subsequently characterizing stromal cell-induced metabolic changes in tumors using noninvasive molecular imaging techniques.
Collapse
Affiliation(s)
- Alina Doctor
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Markus Laube
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Sebastian Meister
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Oliver C. Kiss
- Department of Targetry, Target Chemistry and Radiopharmacy, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, Partner Site Dresden, University Cancer Center (UCC), Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - Sandra Hauser
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany; (A.D.); (M.L.); (S.M.); (K.K.); (S.H.)
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
3
|
McDaniel A, Rothstein KV, Gonzalez D, Nuccitelli R. Nano-Pulse Stimulation Treatment Inhibits Pan02 Murine Pancreatic Tumor Growth and Induces a Long-Term Adaptive Immune Response with Abscopal Effects When Combined with Immune-Enhancing Agents. Bioelectricity 2024; 6:108-117. [PMID: 39119566 PMCID: PMC11304879 DOI: 10.1089/bioe.2024.0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024] Open
Abstract
Pancreatic cancer is associated with a poor prognosis and immunotherapy alone has not demonstrated sufficient efficacy in the treatment of nonresectable tumors. Nano-Pulse Stimulation™ therapy (NPS™) applies nanosecond electric pulses that lead to regulated cell death, exposing tumor antigen to the immune system. To establish a primary Pan02 tumor, mice were intradermally injected with Pan02 cells into the right flank. Secondary, rechallenge tumors and distal, secondary tumors (abscopal response) were established by injecting Pan02 cells into the opposite left flank. After 5 days of tumor growth, one of the tumors was treated with NPS, followed by injection with an immune-enhancing agent to stimulate an immune response. Growth of the treated primary tumor and untreated rechallenge tumors (injected 60-days post-treatment) or distal secondary tumors (injected simultaneously with the primary) was monitored. NPS in combination with the adjuvant and TLR agonist, resiquimod (RES), was the optimal treatment regimen for both eliminating a primary Pan02 tumor as well as inhibiting growth of a Pan02 cell rechallenge tumor. This inhibition of the rechallenge tumor injected 2 months after eliminating the primary tumor suggests a long-term immune response had been stimulated. Additional support for this came from the observations that depleting CD8+ T-cells reduced inhibition of rechallenge tumor growth by 35% and rechallenge tumors had 3-fold more CD8+ T-cells than tumors injected after surgical resection of the primary tumor. When the NPS-treated tumor was immediately injected with the anti-OX40 antibody to agonize the function of the costimulatory T cell receptor, OX40, up to 80% of untreated abscopal tumors were eliminated. NPS plus RES was the most effective at both eliminating a primary tumor and inhibiting a rechallenge tumor. NPS treatment followed by injection of aOX40 was the most effective at inhibiting the growth of an untreated abscopal tumor.
Collapse
Affiliation(s)
- Amanda McDaniel
- Biology Department, Pulse Biosciences, Inc., Hayward, California, USA
| | | | - Dacia Gonzalez
- Biology Department, Pulse Biosciences, Inc., Hayward, California, USA
| | | |
Collapse
|
4
|
Gu A, Li J, Qiu S, Hao S, Yue ZY, Zhai S, Li MY, Liu Y. Pancreatic cancer environment: from patient-derived models to single-cell omics. Mol Omics 2024; 20:220-233. [PMID: 38414408 DOI: 10.1039/d3mo00250k] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Pancreatic cancer (PC) is a highly malignant cancer characterized by poor prognosis, high heterogeneity, and intricate heterocellular systems. Selecting an appropriate experimental model for studying its progression and treatment is crucial. Patient-derived models provide a more accurate representation of tumor heterogeneity and complexity compared to cell line-derived models. This review initially presents relevant patient-derived models, including patient-derived xenografts (PDXs), patient-derived organoids (PDOs), and patient-derived explants (PDEs), which are essential for studying cell communication and pancreatic cancer progression. We have emphasized the utilization of these models in comprehending intricate intercellular communication, drug responsiveness, mechanisms underlying tumor growth, expediting drug discovery, and enabling personalized medical approaches. Additionally, we have comprehensively summarized single-cell analyses of these models to enhance comprehension of intercellular communication among tumor cells, drug response mechanisms, and individual patient sensitivities.
Collapse
Affiliation(s)
- Ao Gu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Jiatong Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Shimei Qiu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - Shenglin Hao
- Department of Functional Neurosurgery, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Zhu-Ying Yue
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Shuyang Zhai
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Meng-Yao Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| | - Yingbin Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China.
| |
Collapse
|
5
|
Elechalawar CK, Gulla SK, Roy RV, Means N, Zhang Y, Asifa S, Robertson DJ, Xu C, Bhattacharya R, Mukherjee P. Biodistribution and therapeutic efficacy of a gold nanoparticle-based targeted drug delivery system against pancreatic cancer. Cancer Lett 2024; 589:216810. [PMID: 38494151 PMCID: PMC11793163 DOI: 10.1016/j.canlet.2024.216810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/22/2024] [Accepted: 03/07/2024] [Indexed: 03/19/2024]
Abstract
Pancreatic cancer is characterized by desmoplasia; crosstalk between pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs) leads to the deposition of extracellular matrix proteins in the tumor environment resulting in poor vascularity. Targeting either PCCs or PSCs individually has produced mixed results, and there is currently no effective strategy to target both cell types simultaneously. Previously, we demonstrated, through in vitro cell culture experiments, that a specific gold nanoparticle-based nanoformulation containing the anti-EGFR antibody cetuximab (C225) as a targeting agent and gemcitabine as a chemotherapeutic agent effectively targets both PCCs and PSCs simultaneously. Herein, we extend our studies to test the ability of these in vitro tested nano formulations to inhibit tumor growth in an orthotopic co-implantation model of pancreatic cancer in vivo. Orthotopic tumors were established by co-implantation of equal numbers of PCCs and PSCs in the mouse pancreas. Among the various formulations tested, 5 nm gold nanoparticles coated with gemcitabine, cetuximab and poly-ethylene glycol (PEG) of molecular weight 1000 Da, which we named ACGP441000, demonstrated optimal efficacy in inhibiting tumor growth. The current study reveals an opportunity to target PCCs and PSCs simultaneously, by exploiting their overexpression of EGFR as a target, in order to inhibit pancreatic cancer growth.
Collapse
Affiliation(s)
- Chandra Kumar Elechalawar
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Suresh Kumar Gulla
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Ram Vinod Roy
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Nicolas Means
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Yushan Zhang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Sima Asifa
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - David J Robertson
- Department of Chemistry and University of Missouri Research Reactor, University of Missouri, Columbia, MO 65211, USA
| | - Chao Xu
- Department of Biostatistics and Epidemiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
6
|
Dutta D, Ray P, De A, Ghosh A, Hazra RS, Ghosh P, Banerjee S, Diaz FJ, Upadhyay SP, Quadir M, Banerjee SK. pH-responsive targeted nanoparticles release ERK-inhibitor in the hypoxic zone and sensitize free gemcitabine in mutant K-Ras-addicted pancreatic cancer cells and mouse model. PLoS One 2024; 19:e0297749. [PMID: 38687749 PMCID: PMC11060587 DOI: 10.1371/journal.pone.0297749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/12/2024] [Indexed: 05/02/2024] Open
Abstract
Therapeutic options for managing Pancreatic ductal adenocarcinoma (PDAC), one of the deadliest types of aggressive malignancies, are limited and disappointing. Therefore, despite suboptimal clinical effects, gemcitabine (GEM) remains the first-line chemotherapeutic drug in the clinic for PDAC treatment. The therapeutic limitations of GEM are primarily due to poor bioavailability and the development of chemoresistance resulting from the addiction of mutant-K-RAS/AKT/ERK signaling-mediated desmoplastic barriers with a hypoxic microenvironment. Several new therapeutic approaches, including nanoparticle-assisted drug delivery, are being investigated by us and others. This study used pH-responsive nanoparticles encapsulated ERK inhibitor (SCH772984) and surface functionalized with tumor-penetrating peptide, iRGD, to target PDAC tumors. We used a small molecule, SCH772984, to target ERK1 and ERK2 in PDAC and other cancer cells. This nanocarrier efficiently released ERKi in hypoxic and low-pH environments. We also found that the free-GEM, which is functionally weak when combined with nanoencapsulated ERKi, led to significant synergistic treatment outcomes in vitro and in vivo. In particular, the combination approaches significantly enhanced the GEM effect in PDAC growth inhibition and prolonged survival of the animals in a genetically engineered KPC (LSL-KrasG12D/+/LSL-Trp53R172H/+/Pdx-1-Cre) pancreatic cancer mouse model, which is not observed in a single therapy. Mechanistically, we anticipate that the GEM efficacy was increased as ERKi blocks desmoplasia by impairing the production of desmoplastic regulatory factors in PDAC cells and KPC mouse tumors. Therefore, 2nd generation ERKi (SCH 772984)-iRGD-pHNPs are vital for the cellular response to GEM and denote a promising therapeutic target in PDAC with mutant K-RAS.
Collapse
Affiliation(s)
- Debasmita Dutta
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Priyanka Ray
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Archana De
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
| | - Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Raj Shankar Hazra
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Pratyusha Ghosh
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Francisco J. Diaz
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Sunil P. Upadhyay
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| | - Mohiuddin Quadir
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, United States of America
| | - Sushanta K. Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, MO, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, United States of America
| |
Collapse
|
7
|
Schmidt P, Lindemeyer J, Raut P, Schütz M, Saniternik S, Jönsson J, Endepols H, Fischer T, Quaas A, Schlößer HA, Thelen M, Grüll H. Multiparametric Characterization of the DSL-6A/C1 Pancreatic Cancer Model in Rats. Cancers (Basel) 2024; 16:1535. [PMID: 38672617 PMCID: PMC11049193 DOI: 10.3390/cancers16081535] [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: 03/05/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The DSL-6A/C1 murine pancreatic ductal adenocarcinoma (PDAC) tumor model was established in Lewis rats and characterized through a comprehensive multiparametric analysis to compare it to other preclinical tumor models and explore potential diagnostic and therapeutical targets. DSL-6A/C1 tumors were histologically analyzed to elucidate PDAC features. The tumor microenvironment was studied for immune cell prevalence. Multiparametric MRI and PET imaging were utilized to characterize tumors, and 68Ga-FAPI-46-targeting cancer-associated fibroblasts (CAFs), were used to validate the histological findings. The histology confirmed typical PDAC characteristics, such as malformed pancreatic ductal malignant cells and CAFs. Distinct immune landscapes were identified, revealing an increased presence of CD8+ T cells and a decreased CD4+ T cell fraction within the tumor microenvironment. PET imaging with 68Ga-FAPI tracers exhibited strong tracer uptake in tumor tissues. The MRI parameters indicated increasing intralesional necrosis over time and elevated contrast media uptake in vital tumor areas. We have demonstrated that the DSL-6A/C1 tumor model, particularly due to its high tumorigenicity, tumor size, and 68Ga-FAPI-46 sensitivity, is a suitable alternative to established small animal models for many forms of preclinical analyses and therapeutic studies of PDAC.
Collapse
Affiliation(s)
- Patrick Schmidt
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Johannes Lindemeyer
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Pranali Raut
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Markus Schütz
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Sven Saniternik
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| | - Jannika Jönsson
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
| | - Heike Endepols
- Faculty of Medicine and University Hospital of Cologne, Institute of Radiochemistry and Experimental Molecular Imaging, University of Cologne, 50937 Cologne, Germany;
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
- Nuclear Chemistry, Institute of Neuroscience and Medicine (INM-5), Forschungszentrum Jülich GmbH, 52425 Jülich, Germany
| | - Thomas Fischer
- Faculty of Medicine and University Hospital of Cologne, Department of Nuclear Medicine, University of Cologne, 50937 Cologne, Germany;
| | - Alexander Quaas
- Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, 50937 Cologne, Germany;
| | - Hans Anton Schlößer
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
- Department of General, Visceral, Cancer and Transplantation Surgery, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Martin Thelen
- Center for Molecular Medicine Cologne, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; (H.A.S.); (M.T.)
| | - Holger Grüll
- Faculty of Medicine and University Hospital of Cologne, Institute of Diagnostic and Interventional Radiology, University of Cologne, 50937 Cologne, Germany; (P.S.); (J.L.); (P.R.); (M.S.); (S.S.); (J.J.)
- Faculty of Mathematics and Natural Sciences, Department of Chemistry, University of Cologne, 50937 Cologne, Germany
| |
Collapse
|
8
|
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
|
9
|
Hadjigeorgiou AG, Stylianopoulos T. Evaluation of growth-induced, mechanical stress in solid tumors and spatial association with extracellular matrix content. Biomech Model Mechanobiol 2023; 22:1625-1643. [PMID: 37129689 DOI: 10.1007/s10237-023-01716-3] [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: 10/14/2022] [Accepted: 03/27/2023] [Indexed: 05/03/2023]
Abstract
Mechanical stresses in solid tumors play an important role in tumor progression and treatment efficacy but their quantification is under-investigated. Here, we developed an experimental and computational approach to calculate growth-induced, residual stresses and applied it to the breast (4T1), pancreatic (PAN02), and fibrosarcoma (MCA205) tumor models. Following resection, tumors are embedded in agarose gels and cuts are made in two perpendicular directions to release residual stress. With the use of image processing, the detailed bulging displacement profile is measured and finite elements models of the bulging geometry are developed for the quantification of the stress levels. The mechanical properties of the tumors are measured in vivo prior to resection with shear wave elastography. We find that the average magnitude of residual stresses ranges from 3.31 to 10.88 kPa, and they are non-uniformly distributed within the tissue due to the heterogeneity of the tumor microenvironment. Interestingly, we demonstrate that a second cut can still release a significant amount of stresses. We further find a strong association of spatial hyaluronan and collagen content with the spatial profile of stress for the MCA205 and PAN02 tumors and a partial association for the 4T1. Interestingly the colocalization of hyaluronan and collagen content had a stronger association with the spatial profile of stress for MCA205, PAN02, and 4T1. Finally, measurements of the elastic modulus with shear wave elastography show a nonlinear correlation with tumor volume for the more fibrotic MCA205 and 4T1 tumors. Overall, our results provide insights for a better understanding of the mechanical behavior of tumors.
Collapse
Affiliation(s)
- Andreas G Hadjigeorgiou
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
10
|
de Barros NR, Gomez A, Ermis M, Falcone N, Haghniaz R, Young P, Gao Y, Aquino AF, Li S, Niu S, Chen R, Huang S, Zhu Y, Eliahoo P, Sun A, Khorsandi D, Kim J, Kelber J, Khademhosseini A, Kim HJ, Li B. Gelatin methacryloyl and Laponite bioink for 3D bioprinted organotypic tumor modeling. Biofabrication 2023; 15:10.1088/1758-5090/ace0db. [PMID: 37348491 PMCID: PMC10683563 DOI: 10.1088/1758-5090/ace0db] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/22/2023] [Indexed: 06/24/2023]
Abstract
Three-dimensional (3D)in vitrotumor models that can capture the pathophysiology of human tumors are essential for cancer biology and drug development. However, simulating the tumor microenvironment is still challenging because it consists of a heterogeneous mixture of various cellular components and biological factors. In this regard, current extracellular matrix (ECM)-mimicking hydrogels used in tumor tissue engineering lack physical interactions that can keep biological factors released by encapsulated cells within the hydrogel and improve paracrine interactions. Here, we developed a nanoengineered ion-covalent cross-linkable bioink to construct 3D bioprinted organotypic tumor models. The bioink was designed to implement the tumor ECM by creating an interpenetrating network composed of gelatin methacryloyl (GelMA), a light cross-linkable polymer, and synthetic nanosilicate (Laponite) that exhibits a unique ionic charge to improve retention of biological factors released by the encapsulated cells and assist in paracrine signals. The physical properties related to printability were evaluated to analyze the effect of Laponite hydrogel on bioink. Low GelMA (5%) with high Laponite (2.5%-3.5%) composite hydrogels and high GelMA (10%) with low Laponite (1.0%-2.0%) composite hydrogels showed acceptable mechanical properties for 3D printing. However, a low GelMA composite hydrogel with a high Laponite content could not provide acceptable cell viability. Fluorescent cell labeling studies showed that as the proportion of Laponite increased, the cells became more aggregated to form larger 3D tumor structures. Reverse transcription-polymerase chain reaction (RT-qPCR) and western blot experiments showed that an increase in the Laponite ratio induces upregulation of growth factor and tissue remodeling-related genes and proteins in tumor cells. In contrast, cell cycle and proliferation-related genes were downregulated. On the other hand, concerning fibroblasts, the increase in the Laponite ratio indicated an overall upregulation of the mesenchymal phenotype-related genes and proteins. Our study may provide a rationale for using Laponite-based hydrogels in 3D cancer modeling.
Collapse
Affiliation(s)
- Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Alejandro Gomez
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Biology, California State University, Northridge, CA 91330, United States of America
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Department of Biology, Baylor University, 101 Bagby Ave, TX 76706, United Ustates of America
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Patric Young
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Yaqi Gao
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Albert-Fred Aquino
- Department of Biology, California State University, Northridge, CA 91330, United States of America
| | - Siyuan Li
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- METU Center of Excellence in Biomaterials and Tissue Engineering, Middle East Technical University, Ankara 06800, Turkey
| | - Siyi Niu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- Department of Biomedical Engineering, Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC 27101, United States of America
| | - RunRun Chen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Shuyi Huang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Payam Eliahoo
- Department of Biology, University of California, Irvine, CA 92697, United States of America
| | - Arthur Sun
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Jinjoo Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Jonathan Kelber
- Department of Biology, California State University, Northridge, CA 91330, United States of America
- Department of Integrative Biology, University of California, Berkeley, CA 94720, United States of America
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- College of Pharmacy, Korea University, Sejong 30019, Republic of Korea
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, CA 90024, United States of America
- Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, United States of America
| |
Collapse
|
11
|
Tunable hybrid hydrogels with multicellular spheroids for modeling desmoplastic pancreatic cancer. Bioact Mater 2023; 25:360-373. [PMID: 36879666 PMCID: PMC9984297 DOI: 10.1016/j.bioactmat.2023.02.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/17/2023] Open
Abstract
The tumor microenvironment consists of diverse, complex etiological factors. The matrix component of pancreatic ductal adenocarcinoma (PDAC) plays an important role not only in physical properties such as tissue rigidity but also in cancer progression and therapeutic responsiveness. Although significant efforts have been made to model desmoplastic PDAC, existing models could not fully recapitulate the etiology to mimic and understand the progression of PDAC. Here, two major components in desmoplastic pancreatic matrices, hyaluronic acid- and gelatin-based hydrogels, are engineered to provide matrices for tumor spheroids composed of PDAC and cancer-associated fibroblasts (CAF). Shape analysis profiles reveals that incorporating CAF contributes to a more compact tissue formation. Higher expression levels of markers associated with proliferation, epithelial to mesenchymal transition, mechanotransduction, and progression are observed for cancer-CAF spheroids cultured in hyper desmoplastic matrix-mimicking hydrogels, while the trend can be observed when those are cultured in desmoplastic matrix-mimicking hydrogels with the presence of transforming growth factor-β1 (TGF-β1). The proposed multicellular pancreatic tumor model, in combination with proper mechanical properties and TGF-β1 supplement, makes strides in developing advanced pancreatic models for resembling and monitoring the progression of pancreatic tumors, which could be potentially applicable for realizing personalized medicine and drug testing applications.
Collapse
|
12
|
Palma AM, Bushnell GG, Wicha MS, Gogna R. Tumor microenvironment interactions with cancer stem cells in pancreatic ductal adenocarcinoma. Adv Cancer Res 2023; 159:343-372. [PMID: 37268400 PMCID: PMC11218813 DOI: 10.1016/bs.acr.2023.02.007] [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] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer in the United States. Additionally, the low survival rate makes PDAC the third-leading cause of cancer-related mortality in the United States, and it is projected that by 2030, it will become the second-leading cause of cancer mortality. Several biological factors contribute to PDAC aggressiveness, and their understanding will narrow the gap from biology to clinical care of PDAC, leading to earlier diagnoses and the development of better treatment options. In this review, we describe the origins of PDAC highlighting the role of cancer stem cells (CSC). CSC, also known as tumor initiating cells, which exhibit a unique metabolism that allows them to maintain a highly plastic, quiescent, immune- and therapy-evasive state. However, CSCs can exit quiescence during proliferation and differentiation, with the capacity to form tumors while constituting a small population in tumor tissues. Tumorigenesis depends on the interactions between CSCs and other cellular and non-cellular components in the microenvironment. These interactions are fundamental to support CSC stemness and are maintained throughout tumor development and metastasis. PDAC is characterized by a massive desmoplastic reaction, which result from the deposition of high amounts of extracellular matrix components by stromal cells. Here we review how this generates a favorable environment for tumor growth by protecting tumor cells from immune responses and chemotherapy and inducing tumor cell proliferation and migration, leading to metastasis formation ultimately leading to death. We emphasize the interactions between CSCs and the tumor microenvironment leading to metastasis formation and posit that better understanding and targeting of these interactions will improve patient outcomes.
Collapse
Affiliation(s)
| | - Grace G Bushnell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Max S Wicha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States.
| | - Rajan Gogna
- VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
13
|
Vudatha V, Herremans KM, Freudenberger DC, Liu C, Trevino JG. In vivo models of pancreatic ductal adenocarcinoma. Adv Cancer Res 2023; 159:75-112. [PMID: 37268402 DOI: 10.1016/bs.acr.2023.02.002] [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] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy with high mortality rate. Within the next decade, PDAC is projected to become the second leading cause of cancer-associated death in the United States. Understanding the pathophysiology of PDAC tumorigenesis and metastases is crucial toward developing new therapeutics. One of the challenges in cancer research is generating in vivo models that closely recapitulate the genomic, histological, and clinical characteristics of human tumors. An ideal model for PDAC not only captures the tumor and stromal environment of human disease, but also allows for mutational control and is easy to reproduce in terms of time and cost. In this review, we highlight evolution of in vivo models for PDAC including spontaneous tumors models (i.e., chemical induction, genetic modification, viral delivery), implantation models including patient derived xenografts (PDX), and humanized PDX. We discuss the implementation of each system and evaluate the benefits and shortcomings of these models. Overall, this review provides a broad overview of prior and current techniques of in vivo PDAC modeling and their associated challenges.
Collapse
Affiliation(s)
- Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Kelly M Herremans
- Department of Surgery, University of Florida College of Medicine, Gainesville, FL, United States
| | - Devon C Freudenberger
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Christopher Liu
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Jose G Trevino
- Department of Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA, United States; Division of Surgical Oncology, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
14
|
Effect of acoustic cluster therapy (ACT®) combined with chemotherapy in a patient-derived xenograft mouse model of pancreatic cancer. J Control Release 2022; 352:1134-1143. [PMID: 36372388 DOI: 10.1016/j.jconrel.2022.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/06/2022] [Accepted: 11/07/2022] [Indexed: 11/23/2022]
Abstract
Pancreatic ductal adenocarcinomas respond poorly to chemotherapy, in part due to the dense tumor stroma that hinders drug delivery. Ultrasound (US) in combination with microbubbles has previously shown promise as a means to improve drug delivery, and the therapeutic efficacy of ultrasound-mediated drug delivery is currently being evaluated in multiple clinical trials. However, most of these utilize echogenic contrast agents engineered for imaging, which might not be optimal compared to specialized formulations tailored for drug delivery. In this study, we evaluated the in vivo efficacy of phase-shifting microbubble-microdroplet clusters that, upon insonation, form bubbles in the size range of 20-30 μm. We developed a patient-derived xenograft model of pancreatic cancer implanted in mice that largely retained the stromal content of the originating tumor and compared tumor growth in mice given chemotherapeutics (nab-paclitaxel plus gemcitabine or liposomal irinotecan) with mice given the same chemotherapeutics in addition to ultrasound and acoustic cluster therapy. We found that acoustic cluster therapy significantly improved the effect of both chemotherapeutic regimens and resulted in 7.2 times higher odds of complete remission of the tumor compared to the chemotherapeutics alone.
Collapse
|
15
|
Gautam SK, Dalal V, Sajja BR, Gupta S, Gulati M, Dwivedi NV, Aithal A, Cox JL, Rachagani S, Liu Y, Chung V, Salgia R, Batra SK, Jain M. Endothelin-axis antagonism enhances tumor perfusion in pancreatic cancer. Cancer Lett 2022; 544:215801. [PMID: 35732216 PMCID: PMC10198578 DOI: 10.1016/j.canlet.2022.215801] [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: 03/14/2022] [Revised: 06/02/2022] [Accepted: 06/16/2022] [Indexed: 11/20/2022]
Abstract
Delivery of therapeutic agents in pancreatic cancer (PC) is impaired due to its hypovascular and desmoplastic tumor microenvironment. The Endothelin (ET)-axis is the major regulator of vasomotor tone under physiological conditions and is highly upregulated in multiple cancers. We investigated the effect of dual endothelin receptor antagonist bosentan on perfusion and macromolecular transport in a PC cell-fibroblast co-implantation tumor model using Dynamic Contrast-Enhanced Magnetic Resonance Imaging (DCE-MRI). Following bosentan treatment, the contrast enhancement ratio and wash-in rates in tumors were two- and nine times higher, respectively, compared to the controls, whereas the time to peak was significantly shorter (7.29 ± 1.29 min v/s 22.08 ± 5.88 min; p = 0.04). Importantly, these effects were tumor selective as the magnitudes of change for these parameters were much lower in muscles. Bosentan treatment also reduced desmoplasia and improved intratumoral distribution of high molecular weight FITC-dextran. Overall, these findings support that targeting the ET-axis can serve as a potential strategy to selectively enhance tumor perfusion and improve the delivery of therapeutic agents in pancreatic tumors.
Collapse
Affiliation(s)
- Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Vipin Dalal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Balasrinivasa R Sajja
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Suprit Gupta
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mansi Gulati
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Nidhi V Dwivedi
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yutong Liu
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Vincent Chung
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology and Experimental Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA, 91010, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
16
|
Targeting PI3K/AKT/mTOR Signaling Pathway in Pancreatic Cancer: From Molecular to Clinical Aspects. Int J Mol Sci 2022; 23:ijms231710132. [PMID: 36077529 PMCID: PMC9456549 DOI: 10.3390/ijms231710132] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
Although pancreatic cancer (PC) was considered in the past an orphan cancer type due to its low incidence, it may become in the future one of the leading causes of cancer death. Pancreatic ductal adenocarcinoma (PDAC) is the most frequent type of PC, being a highly aggressive malignancy and having a 5-year survival rate of less than 10%. Non-modifiable (family history, age, genetic susceptibility) and modifiable (smoking, alcohol, acute and chronic pancreatitis, diabetes mellitus, intestinal microbiota) risk factors are involved in PC pathogenesis. Chronic inflammation induced by various factors plays crucial roles in PC development from initiation to metastasis. In multiple malignant conditions such as PC, cytokines, chemokines, and growth factors activate the class I phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) (PI3K/AKT/mTOR) signaling pathway, which plays key roles in cell growth, survival, proliferation, metabolism, and motility. Currently, mTOR, AKT, and PI3K inhibitors are used in clinical studies. Moreover, PI3K/mTOR dual inhibitors are being tested in vitro and in vivo with promising results for PC patients. The main aim of this review is to present PC incidence, risk factors, tumor microenvironment development, and PI3K/AKT/mTOR dysregulation and inhibitors used in clinical, in vivo, and in vitro studies.
Collapse
|
17
|
Monteiro MV, Ferreira LP, Rocha M, Gaspar VM, Mano JF. Advances in bioengineering pancreatic tumor-stroma physiomimetic Biomodels. Biomaterials 2022; 287:121653. [PMID: 35803021 DOI: 10.1016/j.biomaterials.2022.121653] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 01/18/2023]
Abstract
Pancreatic cancer exhibits a unique bioarchitecture and desmoplastic cancer-stoma interplay that governs disease progression, multi-resistance, and metastasis. Emulating the biological features and microenvironment heterogeneity of pancreatic cancer stroma in vitro is remarkably complex, yet highly desirable for advancing the discovery of innovative therapeutics. Diverse bioengineering approaches exploiting patient-derived organoids, cancer-on-a-chip platforms, and 3D bioprinted living constructs have been rapidly emerging in an endeavor to seamlessly recapitulate major tumor-stroma biodynamic interactions in a preclinical setting. Gathering on this, herein we showcase and discuss the most recent advances in bio-assembling pancreatic tumor-stroma models that mimic key disease hallmarks and its desmoplastic biosignature. A reverse engineering perspective of pancreatic tumor-stroma key elementary units is also provided and complemented by a detailed description of biodesign guidelines that are to be considered for improving 3D models physiomimetic features. This overview provides valuable examples and starting guidelines for researchers envisioning to engineer and characterize stroma-rich biomimetic tumor models. All in all, leveraging advanced bioengineering tools for capturing stromal heterogeneity and dynamics, opens new avenues toward generating more predictive and patient-personalized organotypic 3D in vitro platforms for screening transformative therapeutics targeting the tumor-stroma interplay.
Collapse
Affiliation(s)
- Maria V Monteiro
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Luís P Ferreira
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Marta Rocha
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
18
|
Suresh V, Dash P, Suklabaidya S, Murmu KC, Sasmal PK, Jogdand GM, Parida D, Sethi M, Das B, Mohapatra D, Saha S, Prasad P, Satoskar A, Senapati S. MIF confers survival advantage to pancreatic CAFs by suppressing interferon pathway-induced p53-dependent apoptosis. FASEB J 2022; 36:e22449. [PMID: 35839070 DOI: 10.1096/fj.202101953r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 11/11/2022]
Abstract
The presence of activated pancreatic stellate cells (PSCs) in the pancreatic ductal adenocarcinoma (PDAC) microenvironment plays a significant role in cancer progression. Macrophage migration inhibitory factor (MIF) is overexpressed in PDAC tissues and expressed by both cancer and stromal cells. The pathophysiological role of MIF in PDAC-associated fibroblasts or PSCs is yet to be elucidated. Here we report that the PSCs of mouse or cancer-associated fibroblast cells (CAFs) of human expresses MIF and its receptors, whose expression gets upregulated upon LPS or TNF-α stimulation. In vitro functional experiments showed that MIF significantly conferred a survival advantage to CAFs/PSCs upon growth factor deprivation. Genetic or pharmacological inhibition of MIF also corroborated these findings. Further, co-injection of mouse pancreatic cancer cells with PSCs isolated from Mif-/- or Mif+/+ mice confirmed the pro-survival effect of MIF in PSCs and also demonstrated the pro-tumorigenic role of MIF expressed by CAFs in vivo. Differential gene expression analysis and in vitro mechanistic studies indicated that MIF expressed by activated CAFs/PSCs confers a survival advantage to these cells by suppression of interferon pathway induced p53 dependent apoptosis.
Collapse
Affiliation(s)
- Voddu Suresh
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Pujarini Dash
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Sujit Suklabaidya
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Krushna Chandra Murmu
- Regional Centre for Biotechnology, Faridabad, India
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Prakash K Sasmal
- Department of General Surgery, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Gajendra M Jogdand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Deepti Parida
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Manisha Sethi
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Biswajit Das
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Debasish Mohapatra
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
- Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Subha Saha
- Regional Centre for Biotechnology, Faridabad, India
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Punit Prasad
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Abhay Satoskar
- Department of Microbiology, The Ohio State University, Columbus, Ohio, USA
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Department of Cancer Biology, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
19
|
Pion E, Karnosky J, Boscheck S, Wagner BJ, Schmidt KM, Brunner SM, Schlitt HJ, Aung T, Hackl C, Haerteis S. 3D In Vivo Models for Translational Research on Pancreatic Cancer: The Chorioallantoic Membrane (CAM) Model. Cancers (Basel) 2022; 14:cancers14153733. [PMID: 35954398 PMCID: PMC9367548 DOI: 10.3390/cancers14153733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Simple Summary The 5-year overall survival rate for all stages of pancreatic cancer is relatively low at about only 6%. As a result of this exceedingly poor prognosis, new research models are necessary to investigate this highly malignant cancer. One model that has been used extensively for a vast variety of different cancers is the chorioallantoic membrane (CAM) model. It is based on an exceptionally vascularized membrane that develops within fertilized chicken eggs and can be used for the grafting and analysis of tumor tissue. The aim of the study was to summarize already existing works on pancreatic ductal adenocarcinoma (PDAC) and the CAM model. The results were subdivided into different categories that include drug testing, angiogenesis, personalized medicine, modifications of the model, and further developments to help improve the unfavorable prognosis of this disease. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer with adverse outcomes that have barely improved over the last decade. About half of all patients present with metastasis at the time of diagnosis, and the 5-year overall survival rate across all stages is only 6%. Innovative in vivo research models are necessary to combat this cancer and to discover novel treatment strategies. The chorioallantoic membrane (CAM) model represents one 3D in vivo methodology that has been used in a large number of studies on different cancer types for over a century. This model is based on a membrane formed within fertilized chicken eggs that contain a dense network of blood vessels. Because of its high cost-efficiency, simplicity, and versatility, the CAM model appears to be a highly valuable research tool in the pursuit of gaining more in-depth insights into PDAC. A summary of the current literature on the usage of the CAM model for the investigation of PDAC was conducted and subdivided into angiogenesis, drug testing, modifications, personalized medicine, and further developments. On this comprehensive basis, further research should be conducted on PDAC in order to improve the abysmal prognosis of this malignant disease.
Collapse
Affiliation(s)
- Eric Pion
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
| | - Julia Karnosky
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Sofie Boscheck
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
| | - Benedikt J. Wagner
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Katharina M. Schmidt
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Stefan M. Brunner
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Hans J. Schlitt
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, 94469 Deggendorf, Germany
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; (J.K.); (B.J.W.); (K.M.S.); (S.M.B.); (H.J.S.); (C.H.)
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany; (E.P.); (S.B.); (T.A.)
- Correspondence:
| |
Collapse
|
20
|
Ghosh S, Sun B, Jahagirdar D, Luo D, Ortega J, Straubinger RM, Lovell JF. Single-treatment tumor ablation with photodynamic liposomal irinotecan sucrosulfate. Transl Oncol 2022; 19:101390. [PMID: 35290919 PMCID: PMC8918863 DOI: 10.1016/j.tranon.2022.101390] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/21/2022] [Accepted: 03/01/2022] [Indexed: 12/28/2022] Open
Abstract
Irinotecan (IRI) loaded actively into PEGylated liposomes via a sucrosulfate gradient has been approved recently to treat advanced pancreatic cancer. In this study, a similar liposomal composition was developed that includes a low mole fraction (1 mol.%) of porphyrin-phospholipid (PoP), a photosensitizer that stably incorporates into liposomes, to confer light-triggered IRI release. IRI-loaded PoP liposomes containing ammonium sucrosulfate (ASOS) as a complexing agent were more stable in serum compared to liposomes employing the more conventional ammonium sulfate. Without irradiation, PoP IRI liposomes released less than 5% IRI during 8 h of incubation in bovine serum at 37 °C, but released over 90% of the drug within minutes of exposure to red light (665 nm) irradiation. A single treatment with IRI-PoP liposomes and light exposure (15 mg/kg IRI with 250 J/cm2) resulted in tumor eradication in mice bearing either MIA PaCa-2 tumors or low-passage patient-derived tumor xenografts that recapitulate characteristics of the clinical disease. Analogous monotherapies of IRI or photodynamic therapy were ineffective in controlling tumor growth. Enhanced drug uptake could be visualized within laser-treated tumors by direct in situ imaging of irinotecan. Biodistribution analysis of IRI, its active metabolite (SN-38), and major metabolite (SN-38 G) showed that laser treatment significantly increased tumor accumulation of all IRI-derived molecular species. A pharmacokinetic model that hypothesized tumor vasculature permeabilization as the primary reason underlying the increased drug deposition accounted for the enhanced drug influx into tumors.
Collapse
Affiliation(s)
- Sanjana Ghosh
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, United States of America
| | - Boyang Sun
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York, Buffalo, New York, 14260, United States of America
| | - Dushyant Jahagirdar
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Dandan Luo
- CSL Behring LLC, 1020 1st Avenue, King of Prussia, PA, 19406, United States of America
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| | - Robert M Straubinger
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY United States of America
| | - Jonathan F Lovell
- Department of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, NY, 14260, United States of America.
| |
Collapse
|
21
|
Rengaraj A, Bosc L, Machillot P, McGuckin C, Milet C, Forraz N, Paliard P, Barbier D, Picart C. Engineering of a Microscale Niche for Pancreatic Tumor Cells Using Bioactive Film Coatings Combined with 3D-Architectured Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2022; 14:13107-13121. [PMID: 35275488 PMCID: PMC7614000 DOI: 10.1021/acsami.2c01747] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Two-photon polymerization has recently emerged as a promising technique to fabricate scaffolds for three-dimensional (3D) cell culture and tissue engineering. Here, we combined 3D-printed microscale scaffolds fabricated using two-photon polymerization with a bioactive layer-by-layer film coating. This bioactive coating consists of hyaluronic acid and poly(l-lysine) of controlled stiffness, loaded with fibronectin and bone morphogenic proteins 2 and 4 (BMP2 and BMP4) as matrix-bound proteins. Planar films were prepared using a liquid handling robot directly in 96-well plates to perform high-content studies of cellular processes, especially cell adhesion, proliferation, and BMP-induced signaling. The behaviors of two human pancreatic cell lines PANC1 (immortalized) and PAN092 (patient-derived cell line) were systematically compared and revealed important context-specific cell responses, notably in response to film stiffness and matrix-bound BMPs (bBMPs). Fibronectin significantly increased cell adhesion, spreading, and proliferation for both cell types on soft and stiff films; BMP2 increased cell adhesion and inhibited proliferation of PANC1 cells and PAN092 on soft films. BMP4 enhanced cell adhesion and proliferation of PANC1 and showed a bipolar effect on PAN092. Importantly, PANC1 exhibited a strong dose-dependent BMP response, notably for bBMP2, while PAN092 was insensitive to BMPs. Finally, we proved that it is possible to combine a microscale 3D Ormocomp scaffold fabricated using the two-photon polymerization technique with the bioactive film coating to form a microscale tumor tissue and mimic the early stages of metastatic cancer.
Collapse
Affiliation(s)
- Arunkumar Rengaraj
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Lauriane Bosc
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Paul Machillot
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
| | - Colin McGuckin
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Clément Milet
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Nico Forraz
- Cell Therapy Research Institute, CTIBiotech, 5 avenue Lionel Terray, 69330 Meyzieu, France
| | - Philippe Paliard
- Microlight 3D, 5 avenue du Grand Sablon, 38700 La Tronche, France
| | - Denis Barbier
- Microlight 3D, 5 avenue du Grand Sablon, 38700 La Tronche, France
| | - Catherine Picart
- Univ. Grenoble Alpes, INSERM U1292, CEA, CNRS EMR 5000 BRM, IRIG Institute, CEA, Bât C3, 17 rue des Martyrs, 38054, Grenoble, France
- Grenoble Institute of Engineering, CNRS UMR 5628, LMGP, 3 parvis Louis Néel, 38016 Grenoble, France
- Institut Universitaire de France (IUF), Ministère de l’Enseignement Supérieur, de la Recherche et de I’Industrie, 1 rue Descartes, 75 231 Paris Cedex 05, France
| |
Collapse
|
22
|
Ding L, Tang S, Yu A, Wang A, Tang W, Jia H, Oupický D. Nanoemulsion-Assisted siRNA Delivery to Modulate the Nervous Tumor Microenvironment in the Treatment of Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10015-10029. [PMID: 35188730 PMCID: PMC9153289 DOI: 10.1021/acsami.1c21997] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Pancreatic cancer (PC) is a fatal human cancer, whose progression is highly dependent on the nervous tumor microenvironment. In the present study, cationic perfluorocarbon nanoemulsions were employed as an intraperitoneal delivery platform to facilitate the delivery and penetration of a therapeutic small interfering RNA (siRNA) to orthotopic pancreatic tumors. The nanoemulsion was used to silence the expression of the nerve growth factor (NGF) as a way of favorably modulating the tumor-neuronal interactions in pancreatic tumors. The nanoemulsions exhibited deep tumor penetration that was dependent on exocytosis and enhanced NGF gene silencing in vitro and in vivo when compared with control polycation/siRNA polyplexes, leading to the effective and safe suppression of tumor growth in orthotopic PC. Overall, emulsion-assisted delivery of NGF siRNA is a promising treatment approach for PC by targeting the interactions between the tumor cells and the nervous microenvironment.
Collapse
Affiliation(s)
- Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Siyuan Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Ao Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Anlin Wang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - Huizhen Jia
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA
| |
Collapse
|
23
|
Fluvastatin sensitizes pancreatic cancer cells toward radiation therapy and suppresses radiation- and/or TGF-β-induced tumor-associated fibrosis. J Transl Med 2022; 102:298-311. [PMID: 34773069 DOI: 10.1038/s41374-021-00690-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 11/08/2022] Open
Abstract
Pancreatic cancer (PC) is highly resistant to chemo and radiotherapy. Radiation-induced fibrosis (RIF) is a major cause of clinical concern for various malignancies, including PC. In this study, we aimed to evaluate the radiosensitizing and anti-RIF potential of fluvastatin in PC. Short-term viability and clonogenic survival assays were used to evaluate the radiosensitizing potential of fluvastatin in multiple human and murine PC cell lines. The expression of different proteins was analyzed to understand the mechanisms of fluvastatin-mediated radiosensitization of PC cells and its anti-RIF effects in both mouse and human pancreatic stellate cells (PSCs). Finally, these effects of fluvastatin and/or radiation were assessed in an immune-competent syngeneic murine model of PC. Fluvastatin radiosensitized multiple PC cell lines, as well as radioresistant cell lines in vitro, by inhibiting radiation-induced DNA damage repair response. Nonmalignant cells, such as PSCs and NIH3T3 cells, were less sensitive to fluvastatin-mediated radiosensitization than PC cells. Interestingly, fluvastatin suppressed radiation and/or TGF-β-induced activation of PSCs, as well as the fibrogenic properties of these cells in vitro. Fluvastatin considerably augmented the antitumor effect of external radiation therapy and also suppressed intra-tumor RIF in vivo. These findings suggested that along with radiation, fluvastatin co-treatment may be a potential therapeutic approach against PC.
Collapse
|
24
|
Mollica H, Teo YJ, Tan ASM, Tan DZM, Decuzzi P, Pavesi A, Adriani G. A 3D pancreatic tumor model to study T cell infiltration. Biomater Sci 2021; 9:7420-7431. [PMID: 34706370 DOI: 10.1039/d1bm00210d] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The desmoplastic nature of the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment (TME) prevents the infiltration of T cells and the penetration of chemotherapeutic drugs, posing a challenge to the validation of targeted therapies, including T cell immunotherapies. We present an in vitro 3D PDAC-TME model to observe and quantify T cell infiltration across the vasculature. In a three-channel microfluidic device, PDAC cells are cultured in a collagen matrix in the central channel surrounded, on one side, by endothelial cells (ECs) to mimic a blood vessel and, on the opposite side, by pancreatic stellate cells (PSCs) to simulate exocrine pancreas. The migration of T cells toward the tumor is quantified based on their activation state and TME composition. The presence of EC-lining drastically reduces T cell infiltration, confirming the essential role of the vasculature in controlling T cell trafficking. We show that activated T cells migrate ∼50% more than the not-activated ones toward the cancer cells. Correspondingly, in the absence of cancer cells, both activated and not-activated T cells present similar migration toward the PSCs. The proposed approach could help researchers in testing and optimizing immunotherapies for pancreatic cancer.
Collapse
Affiliation(s)
- Hilaria Mollica
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genova, 16163, Italy
| | - Yi Juan Teo
- Singapore Immunology Network, A*STAR, 8A Biomedical Groove, 138648, Singapore.
| | - Alrina Shin Min Tan
- Singapore Immunology Network, A*STAR, 8A Biomedical Groove, 138648, Singapore.
| | - Damien Zhi Ming Tan
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, 138673, Singapore
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, Genova, 16163, Italy
| | - Andrea Pavesi
- Institute of Molecular and Cell Biology, A*STAR, 61 Biopolis Drive, 138673, Singapore
| | - Giulia Adriani
- Singapore Immunology Network, A*STAR, 8A Biomedical Groove, 138648, Singapore. .,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 117583, Singapore
| |
Collapse
|
25
|
De Silva P, Bano S, Pogue BW, Wang KK, Maytin EV, Hasan T. Photodynamic priming with triple-receptor targeted nanoconjugates that trigger T cell-mediated immune responses in a 3D in vitro heterocellular model of pancreatic cancer. NANOPHOTONICS 2021; 10:3199-3214. [PMID: 37485044 PMCID: PMC10361703 DOI: 10.1515/nanoph-2021-0304] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Photodynamic priming (PDP), a collateral effect of photodynamic therapy, can transiently alter the tumor microenvironment (TME) beyond the cytotoxic zone. Studies have demonstrated that PDP increases tumor permeability and modulates immune-stimulatory effects by inducing immunogenic cell death, via the release of damage-associated molecular patterns and tumor-associated antigens. Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest of cancers with a stubborn immunosuppressive TME and a dense stroma, representing a challenge for current molecular targeted therapies often involving macromolecules. We, therefore, tested the hypothesis that PDP's TME modulation will enable targeted therapy and result in immune stimulation. Using triple-receptor-targeted photoimmuno-nanoconjugate (TR-PINs)-mediated PDP, targeting epidermal growth factor receptor, transferrin receptor, and human epidermal growth factor receptor 2 we show light dose-dependent TR-PINs mediated cytotoxicity inhuman PDA Ccells (MIAPaCa-2),co-cultured with human pancreatic cancer-associated fibroblasts (PCAFs) in spheroids. Furthermore, TR-PINs induced the expression of heat shock proteins (Hsp60, Hsp70), Calreticulin, and high mobility group box 1 in a light dose and time-dependent manner.TR-PINs-mediated T cell activation was observed in co-cultures of immune cells with the MIA PaCa-2-PCAF spheroids. Both CD4+ T and CD8+ T cells showed light dose and time-dependant antitumor reactivity by upregulating degranulation marker CD107a and interferon-gamma post-PDP. Substantial tumor cell death in immune cell-spheroid co-cultures by day 3 shows the augmentation by antitumor T cell activation and their ability to recognize tumors for a light dose-dependent kill. These data confirm enhanced destruction of heterogeneous pancreatic spheroids mediated by PDP-induced phototoxicity, TME modulation and increased immunogenicity with targeted nanoconstructs.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Kenneth K. Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Edward V. Maytin
- Departments of Dermatology and Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Tayyaba Hasan
- Corresponding author: Tayyaba Hasan, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, BAR 314A, Boston, MA, 02114, USA; and Division of Health Sciences and Technology, Massachusetts Institute of Technology, Harvard University, Cambridge, MA, 02139, USA,
| |
Collapse
|
26
|
Hayashi H, Uemura N, Zhao L, Matsumura K, Sato H, Shiraishi Y, Baba H. Biological Significance of YAP/TAZ in Pancreatic Ductal Adenocarcinoma. Front Oncol 2021; 11:700315. [PMID: 34395269 PMCID: PMC8358930 DOI: 10.3389/fonc.2021.700315] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal types of cancer. Despite major advances in defining the molecular mutations driving PDAC, this disease remains universally lethal with an overall 5-year survival rate of only about 7–8%. Genetic alterations in PDAC are exemplified by four critical genes (KRAS, TP53, CDKN2A, and SMAD4) that are frequently mutated. Among these, KRAS mutation ranges from 88% to 100% in several studies. Hippo signaling is an evolutionarily conserved network that plays a key role in normal organ development and tissue regeneration. Its core consists of the serine/threonine kinases mammalian sterile 20-like kinase 1 and 2 (MST1/2) and large tumor suppressor 1 and 2. Interestingly, pancreas-specific MST1/2 double knockout mice have been reported to display a decreased pancreas mass. Many of the genes involved in the Hippo signaling pathway are recognized as tumor suppressors, while the Hippo transducers Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are identified as oncogenes. By dephosphorylation, YAP and TAZ accumulate in the nucleus and interact with transcription factors such as TEA domain transcription factor-1, 2, 3, and 4. Dysregulation of Hippo signaling and activation of YAP/TAZ have been recognized in a variety of human solid cancers, including PDAC. Recent studies have elucidated that YAP/TAZ play a crucial role in the induction of acinar-to-ductal metaplasia, an initial step in the progression to PDAC, in genetically engineered mouse models. YAP and TAZ also play a key role in the development of PDAC by both KRAS-dependent and KRAS-independent bypass mechanisms. YAP/TAZ have become extensively studied in PDAC and their biological importance during the development and progression of PDAC has been uncovered. In this review, we summarize the biological significance of a dysregulated Hippo signaling pathway or activated YAP/TAZ in PDAC and propose a role for YAP/TAZ as a therapeutic target.
Collapse
Affiliation(s)
- Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Liu Zhao
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kazuki Matsumura
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroki Sato
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuta Shiraishi
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
27
|
Heinrich MA, Mostafa AMRH, Morton JP, Hawinkels LJAC, Prakash J. Translating complexity and heterogeneity of pancreatic tumor: 3D in vitro to in vivo models. Adv Drug Deliv Rev 2021; 174:265-293. [PMID: 33895214 DOI: 10.1016/j.addr.2021.04.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely aggressive type of cancer with an overall survival rate of less than 7-8%, emphasizing the need for novel effective therapeutics against PDAC. However only a fraction of therapeutics which seemed promising in the laboratory environment will eventually reach the clinic. One of the main reasons behind this low success rate is the complex tumor microenvironment (TME) of PDAC, a highly fibrotic and dense stroma surrounding tumor cells, which supports tumor progression as well as increases the resistance against the treatment. In particular, the growing understanding of the PDAC TME points out a different challenge in the development of efficient therapeutics - a lack of biologically relevant in vitro and in vivo models that resemble the complexity and heterogeneity of PDAC observed in patients. The purpose and scope of this review is to provide an overview of the recent developments in different in vitro and in vivo models, which aim to recapitulate the complexity of PDAC in a laboratory environment, as well to describe how 3D in vitro models can be integrated into drug development pipelines that are already including sophisticated in vivo models. Hereby a special focus will be given on the complexity of in vivo models and the challenges in vitro models face to reach the same levels of complexity in a controllable manner. First, a brief introduction of novel developments in two dimensional (2D) models and ex vivo models is provided. Next, recent developments in three dimensional (3D) in vitro models are described ranging from spheroids, organoids, scaffold models, bioprinted models to organ-on-chip models including a discussion on advantages and limitations for each model. Furthermore, we will provide a detailed overview on the current PDAC in vivo models including chemically-induced models, syngeneic and xenogeneic models, highlighting hetero- and orthotopic, patient-derived tissues (PDX) models, and genetically engineered mouse models. Finally, we will provide a discussion on overall limitations of both, in vitro and in vivo models, and discuss necessary steps to overcome these limitations to reach an efficient drug development pipeline, as well as discuss possibilities to include novel in silico models in the process.
Collapse
Affiliation(s)
- Marcel A Heinrich
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Ahmed M R H Mostafa
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands
| | - Jennifer P Morton
- Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow G61 1QH, UK
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Centre, PO-box 9600, 2300 RC Leiden, the Netherlands
| | - Jai Prakash
- Department of Biomaterials Science and Technology, Section Targeted Therapeutics, Technical Medical Centre, University of Twente, 7500AE Enschede, the Netherlands.
| |
Collapse
|
28
|
Pfeifer E, Burchell JM, Dazzi F, Sarker D, Beatson R. Apoptosis in the Pancreatic Cancer Tumor Microenvironment-The Double-Edged Sword of Cancer-Associated Fibroblasts. Cells 2021; 10:cells10071653. [PMID: 34359823 PMCID: PMC8305815 DOI: 10.3390/cells10071653] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis. This is attributed to the disease already being advanced at presentation and having a particularly aggressive tumor biology. The PDAC tumor microenvironment (TME) is characterized by a dense desmoplastic stroma, dominated by cancer-associated fibroblasts (CAF), extracellular matrix (ECM) and immune cells displaying immunosuppressive phenotypes. Due to the advanced stage at diagnosis, the depletion of immune effector cells and lack of actionable genomic targets, the standard treatment is still apoptosis-inducing regimens such as chemotherapy. Paradoxically, it has emerged that the direct induction of apoptosis of cancer cells may fuel oncogenic processes in the TME, including education of CAF and immune cells towards pro-tumorigenic phenotypes. The direct effect of cytotoxic therapies on CAF may also enhance tumorigenesis. With the awareness that CAF are the predominant cell type in PDAC driving tumorigenesis with various tumor supportive functions, efforts have been made to try to target them. However, efforts to target CAF have, to date, shown disappointing results in clinical trials. With the help of sophisticated single cell analyses it is now appreciated that CAF in PDAC are a heterogenous population with both tumor supportive and tumor suppressive functions. Hence, there remains a debate whether targeting CAF in PDAC is a valid therapeutic strategy. In this review we discuss how cytotoxic therapies and the induction of apoptosis in PDAC fuels oncogenesis by the education of surrounding stromal cells, with a particular focus on the potential pro-tumorigenic outcomes arising from targeting CAF. In addition, we explore therapeutic avenues to potentially avoid the oncogenic effects of apoptosis in PDAC CAF.
Collapse
|
29
|
Ahmad RS, Eubank TD, Lukomski S, Boone BA. Immune Cell Modulation of the Extracellular Matrix Contributes to the Pathogenesis of Pancreatic Cancer. Biomolecules 2021; 11:biom11060901. [PMID: 34204306 PMCID: PMC8234537 DOI: 10.3390/biom11060901] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/07/2021] [Accepted: 06/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a five-year survival rate of only 9%. PDAC is characterized by a dense, fibrotic stroma composed of extracellular matrix (ECM) proteins. This desmoplastic stroma is a hallmark of PDAC, representing a significant physical barrier that is immunosuppressive and obstructs penetration of cytotoxic chemotherapy agents into the tumor microenvironment (TME). Additionally, dense ECM promotes hypoxia, making tumor cells refractive to radiation therapy and alters their metabolism, thereby supporting proliferation and survival. In this review, we outline the significant contribution of fibrosis to the pathogenesis of pancreatic cancer, with a focus on the cross talk between immune cells and pancreatic stellate cells that contribute to ECM deposition. We emphasize the cellular mechanisms by which neutrophils and macrophages, specifically, modulate the ECM in favor of PDAC-progression. Furthermore, we investigate how activated stellate cells and ECM influence immune cells and promote immunosuppression in PDAC. Finally, we summarize therapeutic strategies that target the stroma and hinder immune cell promotion of fibrogenesis, which have unfortunately led to mixed results. An enhanced understanding of the complex interactions between the pancreatic tumor ECM and immune cells may uncover novel treatment strategies that are desperately needed for this devastating disease.
Collapse
Affiliation(s)
- Ramiz S. Ahmad
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
| | - Timothy D. Eubank
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Slawomir Lukomski
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA;
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA; (T.D.E.); (S.L.)
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
30
|
Yan S, Yan J, Liu D, Li X, Kang Q, You W, Zhang J, Wang L, Tian Z, Lu W, Liu W, He W. A nano-predator of pathological MDMX construct by clearable supramolecular gold(I)-thiol-peptide complexes achieves safe and potent anti-tumor activity. Theranostics 2021; 11:6833-6846. [PMID: 34093856 PMCID: PMC8171083 DOI: 10.7150/thno.59020] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/22/2021] [Indexed: 12/11/2022] Open
Abstract
As alternatives to small-molecular proteolysis-targeting chimeras (PROTAC), peptide-based molecular glues (MG) are a broad range of dual-functional ligands that simultaneously bind with targetable proteins and E3 ligases by mimicking proteinprotein interaction (PPI) partners. Methods: Herein, we design a peptide-derived MG to target a tumor-driving protein, MDMX, for degradation, and nanoengineered it into a supramolecular gold(I)-thiol-peptide complex (Nano-MP) to implement the proteolysis recalcitrance, cellular internalization, and glutathione-triggered release. To optimize the tumor targeting, a pH-responsive macromolecule termed polyacryl sulfydryl imidazole (PSI) was synthesized to coat Nano-MP. Results: As expected, Nano-MP@PSI induced the MDMX degradation by ubiquitination and subsequently restored the anti-cancer function of p53 and p73. Nano-MP@PSI revealed potent anti-cancer activities in an orthotopic xenograft mouse model of retinoblastoma by intraocular injection and a patient-derived xenograft model of malignant pancreatic cancer by systemic injection, while maintaining a favorable safety profile and showing a highly favorable clearable profile of excretion from the living body. Conclusion: Collectively, this work not only provided a clinically viable paradigm for the treatment of a wide variety of tumors by multiple administration types, but, more importantly, it bridged the chasm between peptides and PROTACs, and likely reinvigorated the development of peptide-derived proteolysis-targeting chimeras for a great variety of diseases.
Collapse
Affiliation(s)
- Siqi Yan
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Ophthalmology Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jin Yan
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Dan Liu
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiang Li
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Qianyan Kang
- Ophthalmology Department, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Weiming You
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Jinghua Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Lei Wang
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, 45267 OH, USA
| | - Wuyuan Lu
- School of Basic Medicine, Fudan University, Shanghai 20433, China
| | - Wenjia Liu
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Wangxiao He
- Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
- Institute for Stem Cell & Regenerative Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
31
|
Monteiro MV, Gaspar VM, Mendes L, Duarte IF, Mano JF. Stratified 3D Microtumors as Organotypic Testing Platforms for Screening Pancreatic Cancer Therapies. SMALL METHODS 2021; 5:e2001207. [PMID: 34928079 DOI: 10.1002/smtd.202001207] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/19/2021] [Indexed: 06/14/2023]
Abstract
Cancer-associated pancreatic stellate cells installed in periacinar/periductal regions are master players in generating the characteristic biophysical shield found in pancreatic ductal adenocarcinoma (PDAC). Recreating this unique PDAC stromal architecture and its desmoplastic microenvironment in vitro is key to discover innovative treatments. However, this still remains highly challenging to realize. Herein, organotypic 3D microtumors that recapitulate PDAC-stroma spatial bioarchitecture, as well as its biomolecular, metabolic, and desmoplastic signatures, are bioengineered. Such newly engineered platforms, termed stratified microenvironment spheroid models - STAMS - mimic the spatial stratification of cancer-stromal cells, exhibit a reproducible morphology and sub-millimeter size. In culture, 3D STAMS secrete the key molecular biomarkers found in human pancreatic cancer, namely TGF-β, FGF-2, IL-1β, and MMP-9, among others. This is accompanied by an extensive desmoplastic reaction where collagen and glycosaminoglycans (GAGs) de novo deposition is observed. These stratified models also recapitulate the resistance to various chemotherapeutics when compared to standard cancer-stroma random 3D models. Therapeutics resistance is further evidenced upon STAMS inclusion in a tumor extracellular matrix (ECM)-mimetic hydrogel matrix, reinforcing the importance of mimicking PDAC-stroma bioarchitectural features in vitro. The 3D STAMS technology represents a next generation of biomimetic testing platforms with improved potential for advancing high-throughput screening and preclinical validation of innovative pancreatic cancer therapies.
Collapse
Affiliation(s)
- Maria V Monteiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Luís Mendes
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Iola F Duarte
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
32
|
Mallya K, Gautam SK, Aithal A, Batra SK, Jain M. Modeling pancreatic cancer in mice for experimental therapeutics. Biochim Biophys Acta Rev Cancer 2021; 1876:188554. [PMID: 33945847 DOI: 10.1016/j.bbcan.2021.188554] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy that is characterized by early metastasis, low resectability, high recurrence, and therapy resistance. The experimental mouse models have played a central role in understanding the pathobiology of PDAC and in the preclinical evaluation of various therapeutic modalities. Different mouse models with targetable pathological hallmarks have been developed and employed to address the unique challenges associated with PDAC progression, metastasis, and stromal heterogeneity. Over the years, mouse models have evolved from simple cell line-based heterotopic and orthotopic xenografts in immunocompromised mice to more complex and realistic genetically engineered mouse models (GEMMs) involving multi-gene manipulations. The GEMMs, mostly driven by KRAS mutation(s), have been widely accepted for therapeutic optimization due to their high penetrance and ability to recapitulate the histological, molecular, and pathological hallmarks of human PDAC, including comparable precursor lesions, extensive metastasis, desmoplasia, perineural invasion, and immunosuppressive tumor microenvironment. Advanced GEMMs modified to express fluorescent proteins have allowed cell lineage tracing to provide novel insights and a new understanding about the origin and contribution of various cell types in PDAC pathobiology. The syngeneic mouse models, GEMMs, and target-specific transgenic mice have been extensively used to evaluate immunotherapies and study therapy-induced immune modulation in PDAC yielding meaningful results to guide various clinical trials. The emerging mouse models for parabiosis, hepatic metastasis, cachexia, and image-guided implantation, are increasingly appreciated for their high translational significance. In this article, we describe the contribution of various experimental mouse models to the current understanding of PDAC pathobiology and their utility in evaluating and optimizing therapeutic modalities for this lethal malignancy.
Collapse
Affiliation(s)
- Kavita Mallya
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA.
| | - Abhijit Aithal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
33
|
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
|
34
|
Pham TND, Shields MA, Spaulding C, Principe DR, Li B, Underwood PW, Trevino JG, Bentrem DJ, Munshi HG. Preclinical Models of Pancreatic Ductal Adenocarcinoma and Their Utility in Immunotherapy Studies. Cancers (Basel) 2021; 13:cancers13030440. [PMID: 33503832 PMCID: PMC7865443 DOI: 10.3390/cancers13030440] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Immune checkpoint blockade has provided durable clinical responses in a number of human malignancies, but not in patients with pancreatic cancer. Efforts to understand mechanisms of resistance and increase efficacy of immune checkpoint blockade in pancreatic cancer require the use of appropriate preclinical models in the laboratory. Here, we discuss the benefits, caveats, and potentials for improvement of the most commonly used models, including murine-based and patient-derived models. Abstract The advent of immunotherapy has transformed the treatment landscape for several human malignancies. Antibodies against immune checkpoints, such as anti-PD-1/PD-L1 and anti-CTLA-4, demonstrate durable clinical benefits in several cancer types. However, checkpoint blockade has failed to elicit effective anti-tumor responses in pancreatic ductal adenocarcinoma (PDAC), which remains one of the most lethal malignancies with a dismal prognosis. As a result, there are significant efforts to identify novel immune-based combination regimens for PDAC, which are typically first tested in preclinical models. Here, we discuss the utility and limitations of syngeneic and genetically-engineered mouse models that are currently available for testing immunotherapy regimens. We also discuss patient-derived xenograft mouse models, human PDAC organoids, and ex vivo slice cultures of human PDAC tumors that can complement murine models for a more comprehensive approach to predict response and resistance to immunotherapy regimens.
Collapse
Affiliation(s)
- Thao N. D. Pham
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| | - Mario A. Shields
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
| | - Christina Spaulding
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
| | - Daniel R. Principe
- Medical Scientist Training Program, University of Illinois, Chicago, IL 60612, USA;
| | - Bo Li
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Patrick W. Underwood
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - Jose G. Trevino
- Department of Surgery, University of Florida, Gainesville, FL 32611, USA; (P.W.U.); (J.G.T.)
| | - David J. Bentrem
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hidayatullah G. Munshi
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; (M.A.S.); (C.S.)
- Jesse Brown VA Medical Center, Chicago, IL 60612, USA;
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
- Correspondence: (T.N.D.P.); (H.G.M.); Tel.: +1-312-503-0312 (T.N.D.P.); +1-312-503-2301 (H.G.M.)
| |
Collapse
|
35
|
Yu Y, Yang G, Huang H, Fu Z, Cao Z, Zheng L, You L, Zhang T. Preclinical models of pancreatic ductal adenocarcinoma: challenges and opportunities in the era of precision medicine. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:8. [PMID: 33402215 PMCID: PMC7783994 DOI: 10.1186/s13046-020-01787-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an extremely lethal malignancy, with an average 5-year survival rate of 9% (Siegel RL, Miller KD, Jemal A. Ca Cancer J Clin. 2019;69(1):7-34). The steady increase in mortality rate indicates limited efficacy of the conventional regimen. The heterogeneity of PDAC calls for personalized treatment in clinical practice, which requires the construction of a preclinical system for generating patient-derived models. Currently, the lack of high-quality preclinical models results in ineffective translation of novel targeted therapeutics. This review summarizes applications of commonly used models, discusses major difficulties in PDAC model construction and provides recommendations for integrating workflows for precision medicine.
Collapse
Affiliation(s)
- Yiqi Yu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Gang Yang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hua Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Ziyao Fu
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhe Cao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lianfang Zheng
- Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China. .,Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
36
|
Loe AKH, Rao-Bhatia A, Kim JE, Kim TH. Mesenchymal Niches for Digestive Organ Development, Homeostasis, and Disease. Trends Cell Biol 2020; 31:152-165. [PMID: 33349527 DOI: 10.1016/j.tcb.2020.11.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
Mesenchymal-epithelial crosstalk plays a crucial role in organ development and stem cell function. However, the identity of the mesenchymal cells involved in this exchange was unclear. Recent significant advances in single-cell transcriptomics have defined the heterogeneity of these mesenchymal niches. By combining multiomic profiling, animal models, and organoid culture, new studies have not only demonstrated the roles of diverse mesenchymal cell populations but also defined the mechanisms underlying their regulation of niche signals. Focusing on several digestive organs, we describe how similar and diverse mesenchymal cell populations promote organ development and maintain proper stem cell activity, and how the heterogeneity of mesenchymal niches is altered in digestive diseases such as inflammation and cancer.
Collapse
Affiliation(s)
- Adrian Kwan Ho Loe
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Abilasha Rao-Bhatia
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ji-Eun Kim
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tae-Hee Kim
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
37
|
Choi SR, Yang Y, Huang KY, Kong HJ, Flick MJ, Han B. Engineering of biomaterials for tumor modeling. MATERIALS TODAY. ADVANCES 2020; 8:100117. [PMID: 34541484 PMCID: PMC8448271 DOI: 10.1016/j.mtadv.2020.100117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Development of biomaterials mimicking tumor and its microenvironment has recently emerged for the use of drug discovery, precision medicine, and cancer biology. These biomimetic models have developed by reconstituting tumor and stroma cells within the 3D extracellular matrix. The models are recently extended to recapitulate the in vivo tumor microenvironment, including biological, chemical, and mechanical conditions tailored for specific cancer type and its microenvironment. In spite of the recent emergence of various innovative engineered tumor models, many of these models are still early stage to be adapted for cancer research. In this article, we review the current status of biomaterials engineering for tumor models considering three main aspects - cellular engineering, matrix engineering, and engineering for microenvironmental conditions. Considering cancer-specific variability in these aspects, our discussion is focused on pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC). In addition, we further discussed the current challenges and future opportunities to create reliable and relevant tumor models.
Collapse
Affiliation(s)
- Sae Rome Choi
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Yi Yang
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Kai-Yu Huang
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hyun Joon Kong
- Department of Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Matthew J. Flick
- Department of Pathology and Laboratory Medicine, Lineberger Comprehensive Cancer Center, and Blood Research Center, University of North Carolina, Chapel Hill, NC, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
- Weldon School of Biomedical Engineering and Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
38
|
da Silva Filho AF, de Sousa LM, Consonni SR, da Rocha Pitta MG, Carvalho HF, de Melo Rêgo MJB. Galectin-3 Expression in Pancreatic Cell Lines Under Distinct Autophagy-Inducing Stimulus. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2020; 26:1187-1197. [PMID: 33107424 DOI: 10.1017/s1431927620024526] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Hypoxia and nutrient deprivation are responsible for inducing malignant behavior in neoplastic cells. In these conditions, metabolic stress leads the cells to enhance their autophagic flux and to activate key molecules for homeostasis maintenance. Galectin-3 (Gal-3) is upregulated in pancreatic cancer and it is activated under the hypoxic atmosphere. We aimed to analyze the most effective autophagic-inducing conditions in pancreatic ductal adenocarcinoma cells and the effect exerted under these conditions in association with hypoxia on the Gal-3 expression. Gal-3 and the microtubule-associated protein light chain 3 beta (LC3) were accessed through western blot and immunofluorescence. Degradative vacuole quantification was analyzed by transmission electronic microscopy, and inhibition of Gal-3 was performed using siRNA. According to the analyses, the most effective conditions in the inducement of autophagy for PANC-1 and MIA PaCa-2 cells were nutritional deprivation and complete amino acid/glucose deprivation, respectively. PANC-1 cells presented higher Gal-3 when they were submitted to 24 h of nutritional deprivation alone and simultaneously nutritional and oxygen deprivation. Inhibition of Gal-3 causes a decrease of LC3 levels in all experimental conditions. These results confirm that Gal-3 is modulated by microenvironment factors and the possibility of Gal-3 participating in an adaptive response from PDAC cells to extreme conditions.
Collapse
Affiliation(s)
- Antônio Felix da Silva Filho
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| | - Lizandra Maia de Sousa
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Silvio Roberto Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Maira Galdino da Rocha Pitta
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| | - Hernandes Faustino Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Cidade Universitária Zeferino Vaz, Campinas, São Paulo13083-970, Brazil
| | - Moacyr Jesus Barreto de Melo Rêgo
- Immunomodulation and New Therapy Approaches Laboratory (LINAT), Biochemistry Department, Federal University of Pernambuco (UFPE), Cidade Universitária, Recife, Pernambuco50670-901, Brazil
- Laboratório de Imunomodulação e Novas Abordagens Terapêuticas (LINAT), Therapeutic Innovation Research Center- Suelly Galdino (NUPIT-SG), Biochemistry Department, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego, 1235, Cidade Universitária, Recife, Pernambuco50670-901, Brazil
| |
Collapse
|
39
|
Verloy R, Privat-Maldonado A, Smits E, Bogaerts A. Cold Atmospheric Plasma Treatment for Pancreatic Cancer-The Importance of Pancreatic Stellate Cells. Cancers (Basel) 2020; 12:cancers12102782. [PMID: 32998311 PMCID: PMC7601057 DOI: 10.3390/cancers12102782] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/17/2020] [Accepted: 09/26/2020] [Indexed: 01/18/2023] Open
Abstract
Simple Summary This review aims to highlight the potential of cold plasma, the fourth state of matter, as anti-cancer treatment for pancreatic cancer, and the importance of pancreatic stellate cells in the response to this treatment. Currently, a significant lack of basic research on cold plasma considering both pancreatic cancer and stellate cells exists. However, co-cultures of these populations can be advantageous, as they resemble the cell-to-cell interactions occurring in a tumor in response to therapy. Even more, these studies should be performed prior to clinical trials of cold plasma to avoid unforeseen responses to treatment. This review article provides a framework for future research of cold plasma therapies for pancreatic cancer, considering the critical role of pancreatic stellate cells in the disease and treatment outcome. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a lethal disease with low five-year survival rates of 8% by conventional treatment methods, e.g., chemotherapy, radiotherapy, and surgery. PDAC shows high resistance towards chemo- and radiotherapy and only 15–20% of all patients can have surgery. This disease is predicted to become the third global leading cause of cancer death due to its significant rise in incidence. Therefore, the development of an alternative or combinational method is necessary to improve current approaches. Cold atmospheric plasma (CAP) treatments could offer multiple advantages to this emerging situation. The plasma-derived reactive species can induce oxidative damage and a cascade of intracellular signaling pathways, which could lead to cell death. Previous reports have shown that CAP treatment also influences cells in the tumor microenvironment, such as the pancreatic stellate cells (PSCs). These PSCs, when activated, play a crucial role in the propagation, growth and survival of PDAC tumors. However, the effect of CAP on PSCs is not yet fully understood. This review focuses on the application of CAP for PDAC treatment and the importance of PSCs in the response to treatment.
Collapse
Affiliation(s)
- Ruben Verloy
- Plasma Lab for Applications in Sustainability and Medicine-ANTwerp, University of Antwerp, 2610 Wilrijk, Belgium;
- Center for Oncological Research, University of Antwerp, 2610 Wilrijk, Belgium;
- Correspondence: (R.V.); (A.P.-M.); Tel.: +32-3265-2343 (R.V. & A.P.-M.)
| | - Angela Privat-Maldonado
- Plasma Lab for Applications in Sustainability and Medicine-ANTwerp, University of Antwerp, 2610 Wilrijk, Belgium;
- Center for Oncological Research, University of Antwerp, 2610 Wilrijk, Belgium;
- Correspondence: (R.V.); (A.P.-M.); Tel.: +32-3265-2343 (R.V. & A.P.-M.)
| | - Evelien Smits
- Center for Oncological Research, University of Antwerp, 2610 Wilrijk, Belgium;
| | - Annemie Bogaerts
- Plasma Lab for Applications in Sustainability and Medicine-ANTwerp, University of Antwerp, 2610 Wilrijk, Belgium;
| |
Collapse
|
40
|
Su T, Yang B, Gao T, Liu T, Li J. Polymer nanoparticle-assisted chemotherapy of pancreatic cancer. Ther Adv Med Oncol 2020; 12:1758835920915978. [PMID: 32426046 PMCID: PMC7222269 DOI: 10.1177/1758835920915978] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/20/2020] [Indexed: 12/16/2022] Open
Abstract
Pancreatic cancer is a lethal disease characterized by highly dense stroma fibrosis. Only 15-20% of patients with pancreatic cancer have resectable tumors, and only around 20% of them survive to 5 years. Traditional cancer treatments have little effect on their prognosis, and successful surgical resection combined with effective perioperative therapy is the main method for maximizing long-term survival. For this reason, chemotherapy is an adjunct treatment for resectable cancer and is the main therapy for incurable pancreatic cancer, including metastatic pancreatic adenocarcinoma. However, there are various side effects of chemotherapeutic medicine and low drug penetration because the complex tumor microenvironment limits the application of chemotherapy. As a novel strategy, polymer nanoparticles make it possible to target the tumor microenvironment, release cytotoxic agents through various responsive reactions, and thus overcome the treatment barrier. As drug carriers, polymer nanoparticles show marked advantages, such as increased drug delivery and efficiency, controlled drug release, decreased side effects, prolonged half-life, and evasion of immunogenic blockade. In this review, we discuss the factors that cause chemotherapy obstacles in pancreatic cancer, and introduce the application of polymer nanoparticles to treat pancreatic cancer.
Collapse
Affiliation(s)
- Tianqi Su
- Department of General Surgery, The Second Hospital of Jilin University, Changchun, People’s Republic of China
| | - Bo Yang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Tianren Gao
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, People’s Republic of China
| | - Tongjun Liu
- Department of General Surgery, Second Hospital of Jilin University, Changchun 130041, People’s Republic of China
| | - Jiannan Li
- Department of General Surgery, Second Hospital of Jilin University, Changchun 130041, People’s Republic of China
| |
Collapse
|
41
|
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
|
42
|
Thomas D, Radhakrishnan P. Pancreatic Stellate Cells: The Key Orchestrator of The Pancreatic Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1234:57-70. [PMID: 32040855 DOI: 10.1007/978-3-030-37184-5_5] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Pancreatic cancer is one of the most challenging adenocarcinomas due to its hostile molecular behavior and complex tumor microenvironment. It has been recently postulated that pancreatic stellate cells (PSCs), the resident lipid-storing cells of the pancreas, are important components of the tumor microenvironment as they can transdifferentiate into highly proliferative myofibroblasts in the context of tissue injury. Targeting tumor-stromal crosstalk in the tumor microenvironment has emerged as a promising therapeutic strategy against pancreatic cancer progression and metastasis. This chapter brings a broad view on the biological and pathological role of PSCs in the pancreas, activated stellate cells in the onset of tissue fibrosis, and tumor progression with particular emphasis on the bidirectional interactions between tumor cells and PSCs. Further, potential therapeutic regimens targeting activated PSCs in the pre-clinical and clinical trials are discussed.
Collapse
Affiliation(s)
- Divya Thomas
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA. .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
43
|
Al Alawi R, Alhamdani MSS, Hoheisel JD, Baqi Y. Antifibrotic and tumor microenvironment modulating effect of date palm fruit (Phoenix dactylifera L.) extracts in pancreatic cancer. Biomed Pharmacother 2020; 121:109522. [DOI: 10.1016/j.biopha.2019.109522] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 09/22/2019] [Accepted: 10/01/2019] [Indexed: 12/16/2022] Open
|
44
|
Juin A, Spence HJ, Martin KJ, McGhee E, Neilson M, Cutiongco MFA, Gadegaard N, Mackay G, Fort L, Lilla S, Kalna G, Thomason P, Koh YWH, Norman JC, Insall RH, Machesky LM. N-WASP Control of LPAR1 Trafficking Establishes Response to Self-Generated LPA Gradients to Promote Pancreatic Cancer Cell Metastasis. Dev Cell 2019; 51:431-445.e7. [PMID: 31668663 PMCID: PMC6863394 DOI: 10.1016/j.devcel.2019.09.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 07/23/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma is one of the most invasive and metastatic cancers and has a dismal 5-year survival rate. We show that N-WASP drives pancreatic cancer metastasis, with roles in both chemotaxis and matrix remodeling. lysophosphatidic acid, a signaling lipid abundant in blood and ascites fluid, is both a mitogen and chemoattractant for cancer cells. Pancreatic cancer cells break lysophosphatidic acid down as they respond to it, setting up a self-generated gradient driving tumor egress. N-WASP-depleted cells do not recognize lysophosphatidic acid gradients, leading to altered RhoA activation, decreased contractility and traction forces, and reduced metastasis. We describe a signaling loop whereby N-WASP and the endocytic adapter SNX18 promote lysophosphatidic acid-induced RhoA-mediated contractility and force generation by controlling lysophosphatidic acid receptor recycling and preventing degradation. This chemotactic loop drives collagen remodeling, tumor invasion, and metastasis and could be an important target against pancreatic cancer spread.
Collapse
Affiliation(s)
| | | | | | | | | | - Marie F A Cutiongco
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | - Nikolaj Gadegaard
- Division of Biomedical Engineering, School of Engineering, University of Glasgow, Glasgow G12 8LT, UK
| | | | - Loic Fort
- CRUK Beatson Institute, Glasgow G61 1BD, UK
| | | | | | | | | | - Jim C Norman
- CRUK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Robert H Insall
- CRUK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK
| | - Laura M Machesky
- CRUK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, UK.
| |
Collapse
|
45
|
Rozengurt E, Eibl G. Central role of Yes-associated protein and WW-domain-containing transcriptional co-activator with PDZ-binding motif in pancreatic cancer development. World J Gastroenterol 2019; 25:1797-1816. [PMID: 31057295 PMCID: PMC6478619 DOI: 10.3748/wjg.v25.i15.1797] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a deadly disease with no efficacious treatment options. PDAC incidence is projected to increase, which may be caused at least partially by the obesity epidemic. Significantly enhanced efforts to prevent or intercept this cancer are clearly warranted. Oncogenic KRAS mutations are recognized initiating events in PDAC development, however, they are not entirely sufficient for the development of fully invasive PDAC. Additional genetic alterations and/or environmental, nutritional, and metabolic signals, as present in obesity, type-2 diabetes mellitus, and inflammation, are required for full PDAC formation. We hypothesize that oncogenic KRAS increases the intensity and duration of the growth-promoting signaling network. Recent exciting studies from different laboratories indicate that the activity of the transcriptional co-activators Yes-associated protein (YAP) and WW-domain-containing transcriptional co-activator with PDZ-binding motif (TAZ) play a critical role in the promotion and maintenance of PDAC operating as key downstream target of KRAS signaling. While initially thought to be primarily an effector of the tumor-suppressive Hippo pathway, more recent studies revealed that YAP/TAZ subcellular localization and co-transcriptional activity is regulated by multiple upstream signals. Overall, YAP has emerged as a central node of transcriptional convergence in growth-promoting signaling in PDAC cells. Indeed, YAP expression is an independent unfavorable prognostic marker for overall survival of PDAC. In what follows, we will review studies implicating YAP/TAZ in pancreatic cancer development and consider different approaches to target these transcriptional regulators.
Collapse
Affiliation(s)
- Enrique Rozengurt
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| | - Guido Eibl
- Department of Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
- CURE: Digestive Diseases Research Center, Los Angeles, CA 90095, United States
| |
Collapse
|
46
|
Lenggenhager D, Amrutkar M, Sántha P, Aasrum M, Löhr JM, Gladhaug IP, Verbeke CS. Commonly Used Pancreatic Stellate Cell Cultures Differ Phenotypically and in Their Interactions with Pancreatic Cancer Cells. Cells 2019; 8:cells8010023. [PMID: 30621293 PMCID: PMC6356867 DOI: 10.3390/cells8010023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 12/24/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
Activated pancreatic stellate cells (PSCs) play a central role in the tumor stroma of pancreatic ductal adenocarcinoma (PDAC). Given the limited availability of patient-derived PSCs from PDAC, immortalized PSC cell lines of murine and human origin have been established; however, it is not elucidated whether differences in species, organ disease status, donor age, and immortalization alter the PSC phenotype and behavior compared to that of patient-derived primary PSC cultures. Therefore, a panel of commonly used PSC cultures was examined for important phenotypical and functional features: three primary cultures from human PDAC, one primary from normal human pancreas, and three immortalized (one from human, two from murine pancreas). Growth rate was considerably lower in primary PSCs from human PDAC. Basal collagen synthesis varied between the PSC cultures, and TGF-β stimulation increased collagen synthesis only in non-immortalized cultures. Differences in secretome composition were observed along with a divergence in the DNA synthesis, migration, and response to gemcitabine of PDAC cell lines that were grown in conditioned medium from the various PSC cultures. The findings reveal considerable differences in features and functions that are key to PSCs and in the interactions with PDAC. These observations may be relevant to researchers when selecting the most appropriate PSC culture for their experiments.
Collapse
Affiliation(s)
- Daniela Lenggenhager
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316 Oslo, Norway.
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316 Oslo, Norway.
- Department of Pathology and Molecular Pathology, University Hospital Zürich, University of Zürich, Schmelzbergstrasse 12, 8091 Zürich, Switzerland.
| | - Manoj Amrutkar
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316 Oslo, Norway.
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, P.O. Box 1171 Blindern, 0318 Oslo, Norway.
| | - Petra Sántha
- Department of Pathology, Oslo University Hospital Rikshospitalet, Nydalen, 0424 Oslo, Norway.
| | - Monica Aasrum
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316 Oslo, Norway.
| | - Johannes-Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, K 53, 141 86 Stockholm, Sweden.
| | - Ivar P Gladhaug
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, P.O. Box 1171 Blindern, 0318 Oslo, Norway.
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital Rikshospitalet, Nydalen, 0424 Oslo, Norway.
| | - Caroline S Verbeke
- Department of Pathology, Institute of Clinical Medicine, University of Oslo, Blindern, 0316 Oslo, Norway.
- Department of Pathology, Oslo University Hospital Rikshospitalet, Nydalen, 0424 Oslo, Norway.
| |
Collapse
|
47
|
Dash P, Minz AP, Senapati S. Possible Autocrine Function of Galectin-3 in Pancreatic Stellate Cells. Gastroenterology 2018; 155:933-934. [PMID: 30092185 DOI: 10.1053/j.gastro.2018.01.076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 01/26/2018] [Indexed: 12/02/2022]
Affiliation(s)
- Pujarini Dash
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Aliva Prity Minz
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models Lab, Institute of Life Sciences, Bhubaneswar, Odisha, India
| |
Collapse
|
48
|
Ali MS, Gill KS, Saglio G, Cilloni D, Soden DM, Forde PF. Expressional changes in stemness markers post electrochemotherapy in pancreatic cancer cells. Bioelectrochemistry 2018; 122:84-92. [DOI: 10.1016/j.bioelechem.2018.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/14/2018] [Accepted: 03/14/2018] [Indexed: 12/12/2022]
|
49
|
Nattress CB, Halldén G. Advances in oncolytic adenovirus therapy for pancreatic cancer. Cancer Lett 2018; 434:56-69. [PMID: 29981812 DOI: 10.1016/j.canlet.2018.07.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 02/06/2023]
Abstract
Survival rates for pancreatic cancer patients have remained unchanged for the last four decades. The most aggressive, and most common, type of pancreatic cancer is pancreatic ductal adenocarcinoma (PDAC), which has the lowest 5-year survival rate of all cancers globally. The poor prognosis is typically due to late presentation of often non-specific symptoms and rapid development of resistance to all current therapeutics, including the standard-of-care cytotoxic drug gemcitabine. While early surgical intervention can significantly prolong patient survival, there are few treatment options for late-stage non-resectable metastatic disease, resulting in mostly palliative care. In addition, a defining feature of pancreatic cancer is the immunosuppressive and impenetrable desmoplastic stroma that blocks access to tumour cells by therapeutic drugs. The limited effectiveness of conventional chemotherapeutics reveals an urgent need to develop novel therapies with different mechanisms of action for this malignancy. An emerging alternative to current therapeutics is oncolytic adenoviruses; these engineered biological agents have proven efficacy and tumour-selectivity in preclinical pancreatic cancer models, including models of drug-resistant cancer. Safety of oncolytic adenoviral mutants has been extensively assessed in clinical trials with only limited toxicity to normal healthy tissue being reported. Promising efficacy in combination with gemcitabine was demonstrated in preclinical and clinical studies. A recent surge in novel adenoviral mutants entering clinical trials for pancreatic cancer indicates improved efficacy through activation of the host anti-tumour responses. The potential for adenoviruses to synergise with chemotherapeutics, activate anti-tumour immune responses, and contribute to stromal dissemination render these mutants highly attractive candidates for improved patient outcomes. Currently, momentum is gathering towards the development of systemically-deliverable mutants that are able to overcome anti-viral host immune responses, erythrocyte binding and hepatic uptake, to promote elimination of primary and metastatic lesions. This review will cover the key components of pancreatic cancer oncogenesis; novel oncolytic adenoviruses; clinical trials; and the current progress in overcoming the challenges of systemic delivery.
Collapse
Affiliation(s)
- Callum Baird Nattress
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom
| | - Gunnel Halldén
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, United Kingdom.
| |
Collapse
|