101
|
Zhang T, Zhu S, Huang GW. ALKBH5 suppresses autophagic flux via N6-methyladenosine demethylation of ZKSCAN3 mRNA in acute pancreatitis. World J Gastroenterol 2024; 30:1764-1776. [PMID: 38617741 PMCID: PMC11008368 DOI: 10.3748/wjg.v30.i12.1764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/03/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Increasing evidence has demonstrated that N6-methyladenosine (m6A) RNA modification plays an essential role in a wide range of pathological conditions. Impaired autophagy is a critical hallmark of acute pancreatitis (AP). AIM To explore the role of the m6A modification of ZKSCAN3 in the regulation of autophagy in AP. METHODS The AP mouse cell model was established by cerulein-treated mouse pancreatic acinar cells (MPC-83), and the results were confirmed by the levels of amylase and inflammatory factors. Autophagy activity was evaluated by specific identification of the autophagy-related microstructure and the expression of autophagy-related genes. ZKSCAN3 and ALKBH5 were knocked down to study the function in AP. A m6A RNA binding protein immunoprecipitation assay was used to study how the m6A modification of ZKSCAN3 mRNA is regulated by ALKBH. RESULTS The increased expression of amylase and inflammatory factors in the supernatant and the accumulation of autophagic vacuoles verified that the AP mouse cell model was established. The downregulation of LAMP2 and upregulation of LC3-II/I and SQSTM1 demonstrated that autophagy was impaired in AP. The expression of ZKSCAN3 was upregulated in AP. Inhibition of ZKSCAN3 increased the expression of LAMP2 and decreased the expression of the inflammatory factors, LC3-II/I and SQSTM1. Furthermore, ALKBH5 was upregulated in AP. Knockdown of ALKBH5 downregulated ZKSCAN3 expression and restored decreased autophagic flux in AP. Notably, the bioinformatic analysis revealed 23 potential m6A modification sites on ZKSCAN3 mRNA. The m6A modification of ZKSCAN3 mRNA was significantly decreased in AP. Knockdown of ALKBH5 increased the modification of ZKSCAN3 mRNA, which confirmed that ALKBH5 upregulated ZKSCAN3 expression in a m6A-dependent manner. CONCLUSION ALKBH5 inhibits autophagic flux through m6A demethylation of ZKSCAN3 mRNA in AP, thereby aggravating the severity of the disease.
Collapse
Affiliation(s)
- Tao Zhang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
| | - Shuai Zhu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
| | - Geng-Wen Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410005, Hunan, China
| |
Collapse
|
102
|
Zwirner S, Abu Rmilah AA, Klotz S, Pfaffenroth B, Kloevekorn P, Moschopoulou AA, Schuette S, Haag M, Selig R, Li K, Zhou W, Nelson E, Poso A, Chen H, Amiot B, Jia Y, Minshew A, Michalak G, Cui W, Rist E, Longerich T, Jung B, Felgendreff P, Trompak O, Premsrirut PK, Gries K, Muerdter TE, Heinkele G, Wuestefeld T, Shapiro D, Weissbach M, Koenigsrainer A, Sipos B, Ab E, Zacarias MO, Theisgen S, Gruenheit N, Biskup S, Schwab M, Albrecht W, Laufer S, Nyberg S, Zender L. First-in-class MKK4 inhibitors enhance liver regeneration and prevent liver failure. Cell 2024; 187:1666-1684.e26. [PMID: 38490194 PMCID: PMC11011246 DOI: 10.1016/j.cell.2024.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/20/2023] [Accepted: 02/20/2024] [Indexed: 03/17/2024]
Abstract
Diminished hepatocyte regeneration is a key feature of acute and chronic liver diseases and after extended liver resections, resulting in the inability to maintain or restore a sufficient functional liver mass. Therapies to restore hepatocyte regeneration are lacking, making liver transplantation the only curative option for end-stage liver disease. Here, we report on the structure-based development and characterization (nuclear magnetic resonance [NMR] spectroscopy) of first-in-class small molecule inhibitors of the dual-specificity kinase MKK4 (MKK4i). MKK4i increased liver regeneration upon hepatectomy in murine and porcine models, allowed for survival of pigs in a lethal 85% hepatectomy model, and showed antisteatotic and antifibrotic effects in liver disease mouse models. A first-in-human phase I trial (European Union Drug Regulating Authorities Clinical Trials [EudraCT] 2021-000193-28) with the clinical candidate HRX215 was conducted and revealed excellent safety and pharmacokinetics. Clinical trials to probe HRX215 for prevention/treatment of liver failure after extensive oncological liver resections or after transplantation of small grafts are warranted.
Collapse
Affiliation(s)
- Stefan Zwirner
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany; HepaRegeniX GmbH, Tübingen 72072, Germany
| | - Anan A Abu Rmilah
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Sabrina Klotz
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Bent Pfaffenroth
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen 72076, Germany
| | - Philip Kloevekorn
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen 72076, Germany
| | - Athina A Moschopoulou
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Svenja Schuette
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Mathias Haag
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Roland Selig
- HepaRegeniX GmbH, Tübingen 72072, Germany; Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen 72076, Germany
| | - Kewei Li
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Zhou
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Erek Nelson
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Antti Poso
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany; School of Pharmacy, University of Eastern Finland, Kuopio 70211, Finland; iFIT Cluster of Excellence (EXC 2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen 72076, Germany
| | - Harvey Chen
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Bruce Amiot
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Yao Jia
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Anna Minshew
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Gregory Michalak
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Wei Cui
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Elke Rist
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Thomas Longerich
- Institute of Pathology, University Hospital Heidelberg, Heidelberg 69120, Germany
| | | | - Philipp Felgendreff
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA
| | - Omelyan Trompak
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | | | - Katharina Gries
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Thomas E Muerdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Georg Heinkele
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany
| | - Torsten Wuestefeld
- Laboratory for In Vivo Genetics & Gene Therapy, Genome Institute of Singapore, Agency for Science, Technology and Research (A(∗)STAR), Singapore 138672, Singapore; School of Biological Sciences, Nanyang Technological University of Singapore, Singapore 637551, Singapore
| | | | | | - Alfred Koenigsrainer
- iFIT Cluster of Excellence (EXC 2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen 72076, Germany; German Cancer Research Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Department of General-, Visceral, and Transplant Surgery, University Hospital Tübingen, Tübingen 72076, Germany
| | - Bence Sipos
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany
| | - Eiso Ab
- ZoBio B.V., Leiden 2333 CH, the Netherlands
| | | | | | | | | | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart 70376, Germany; iFIT Cluster of Excellence (EXC 2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen 72076, Germany; Department of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tübingen, Tübingen 72076, Germany
| | | | - Stefan Laufer
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen 72076, Germany; Tübingen Center for Academic Drug Discovery & Development (TüCAD(2)), Tübingen 72076, Germany.
| | - Scott Nyberg
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN 55905, USA.
| | - Lars Zender
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tübingen, Tübingen 72076, Germany; iFIT Cluster of Excellence (EXC 2180) "Image-guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen 72076, Germany; German Cancer Research Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Tübingen Center for Academic Drug Discovery & Development (TüCAD(2)), Tübingen 72076, Germany.
| |
Collapse
|
103
|
Lee KE, Tu VY, Faye AS. Optimal Management of Refractory Crohn's Disease: Current Landscape and Future Direction. Clin Exp Gastroenterol 2024; 17:75-86. [PMID: 38558912 PMCID: PMC10981422 DOI: 10.2147/ceg.s359376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
Refractory Crohn's disease, defined as ongoing inflammation despite the trial of multiple advanced therapies, impacts a number of individuals with Crohn's disease, and leads to significant burden in quality of life and cost. Interventions such as early implementation of advanced therapies, optimization of current therapies prior to switching to an alternative, as well as understanding the overlapping pathophysiology between immune-mediated disorders, however, can help shift the current landscape and reduce the number of patients with refractory disease. As such, in this review we summarize the key takeaways of the latest research in the management of moderate-to-severe Crohn's disease, focusing on maximization of our currently available medications, while also exploring topics such as combination advanced therapies. We also describe evidence for emerging and alternative therapeutic modalities, including fecal microbiota transplant, exclusive enteral feeding, hyperbaric oxygen, stem cell therapy, bone marrow transplant, and posaconazole, with a focus on both the potential impact and specific indications for each.
Collapse
Affiliation(s)
- Kate E Lee
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Violet Y Tu
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Adam S Faye
- Department of Gastroenterology, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
104
|
Li CMY, Briggs MT, Lee YR, Tin T, Young C, Pierides J, Kaur G, Drew P, Maddern GJ, Hoffmann P, Klingler-Hoffmann M, Fenix K. Use of tryptic peptide MALDI mass spectrometry imaging to identify the spatial proteomic landscape of colorectal cancer liver metastases. Clin Exp Med 2024; 24:53. [PMID: 38492056 PMCID: PMC10944452 DOI: 10.1007/s10238-024-01311-5] [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: 01/29/2024] [Accepted: 02/22/2024] [Indexed: 03/18/2024]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths worldwide. CRC liver metastases (CRLM) are often resistant to conventional treatments, with high rates of recurrence. Therefore, it is crucial to identify biomarkers for CRLM patients that predict cancer progression. This study utilised matrix-assisted laser desorption/ionisation mass spectrometry imaging (MALDI-MSI) in combination with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to spatially map the CRLM tumour proteome. CRLM tissue microarrays (TMAs) of 84 patients were analysed using tryptic peptide MALDI-MSI to spatially monitor peptide abundances across CRLM tissues. Abundance of peptides was compared between tumour vs stroma, male vs female and across three groups of patients based on overall survival (0-3 years, 4-6 years, and 7+ years). Peptides were then characterised and matched using LC-MS/MS. A total of 471 potential peptides were identified by MALDI-MSI. Our results show that two unidentified m/z values (1589.876 and 1092.727) had significantly higher intensities in tumours compared to stroma. Ten m/z values were identified to have correlation with biological sex. Survival analysis identified three peptides (Histone H4, Haemoglobin subunit alpha, and Inosine-5'-monophosphate dehydrogenase 2) and two unidentified m/z values (1305.840 and 1661.060) that were significantly higher in patients with shorter survival (0-3 years relative to 4-6 years and 7+ years). This is the first study using MALDI-MSI, combined with LC-MS/MS, on a large cohort of CRLM patients to identify the spatial proteome in this malignancy. Further, we identify several protein candidates that may be suitable for drug targeting or for future prognostic biomarker development.
Collapse
Affiliation(s)
- Celine Man Ying Li
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, SA, 5011, Australia
| | - Matthew T Briggs
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Yea-Rin Lee
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - Teresa Tin
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, SA, 5011, Australia
| | - Clifford Young
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | - John Pierides
- SA Pathology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, University Sains Malaysia, 11800, Pulau Pinang, Malaysia
| | - Paul Drew
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, SA, 5011, Australia
| | - Guy J Maddern
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, SA, 5011, Australia
| | - Peter Hoffmann
- Clinical and Health Sciences, University of South Australia, Adelaide, SA, 5000, Australia
| | | | - Kevin Fenix
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia.
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, SA, 5011, Australia.
| |
Collapse
|
105
|
Oryani MA, Nosrati S, Javid H, Mehri A, Hashemzadeh A, Karimi-Shahri M. Targeted cancer treatment using folate-conjugated sponge-like ZIF-8 nanoparticles: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1377-1404. [PMID: 37715816 DOI: 10.1007/s00210-023-02707-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/02/2023] [Indexed: 09/18/2023]
Abstract
ZIF-8 (zeolitic imidazolate framework-8) is a potential drug delivery system because of its unique properties, which include a large surface area, a large pore capacity, a large loading capacity, and outstanding stability under physiological conditions. ZIF-8 nanoparticles may be readily functionalized with targeting ligands for the identification and absorption of particular cancer cells, enhancing the efficacy of chemotherapeutic medicines and reducing adverse effects. ZIF-8 is also pH-responsive, allowing medication release in the acidic milieu of cancer cells. Because of its tunable structure, it can be easily functionalized to design cancer-specific targeted medicines. The delivery of ZIF-8 to cancer cells can be facilitated by folic acid-conjugation. Hence, it can bind to overexpressed folate receptors on the surface of cancer cells, which holds the promise of reducing unwanted deliveries. As a result of its importance in cancer treatment, the folate-conjugated ZIF-8 was the major focus of this review.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shamim Nosrati
- Department of Clinical Biochemistry, Faculty of Medicine, Azad Shahroud University, Shahroud, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Mehri
- Endoscopic and Minimally Invasive Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
106
|
Song Y, Zou W, Gao Y, Zhao Z, Yin Z, Xiao C, Liu Q, Liu R. Short- and long-term outcomes of robotic versus open radical antegrade modular pancreatosplenectomy: a retrospective propensity score-matched cohort study. Surg Endosc 2024; 38:1316-1328. [PMID: 38110793 DOI: 10.1007/s00464-023-10635-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Robotic distal pancreatectomy has increasingly been accepted as it has overcome some of the limitations of open distal pancreatectomy, whilst the outcomes following robotic radical antegrade modular pancreatosplenectomy (RAMPS) in patients with pancreatic ductal adenocarcinoma (PDAC) are still uncertain. This study aimed to evaluate the short and long-term outcomes of robotic RAMPS and open RAMPS for PDAC. METHODS The patients who underwent robotic RAMPS and open RAMPS for PDAC at our clinical centre between January 2017 and December 2021 were reviewed. After a propensity score matching (PSM) at a 1:1 ratio, the perioperative and pathological outcomes in the both groups were reviewed. Univariable and multivariable Cox regression analyses were used to identify independent prognosis factors for overall survival (OS) and recurrence-free survival (RFS) of these patients. RESULTS 318 cases were recorded in robotic and open groups. The robotic group showed advantages in operative time [205.00 (166.00, 240.00) min vs 235 (184.75, 270.00) min, P = 0.002], estimated blood loss [100 (50, 100) ml vs 300 (100, 400) ml, P < 0.001], delayed gastric emptying [0 vs 5.03%, P = 0.007] and postoperative hospital stay [7.00 (5.00, 10.00) days vs 11.00 (8.00, 14.00) days, P < 0.001]. There were no significant differences in rate of severe postoperative complications between the robotic group and the open group. Multivariable analysis showed that carbohydrate antigen 19-9, estimated blood loss, N stage, tumour differentiation, chemotherapy and vascular invasion were independent risk factors for OS and RFS of these patients. CONCLUSIONS Robotic RAMPS was safe and had some advantages over open RAMPS for PDAC. There were no significantly differences in oncological outcomes and long-term survival rates between the robotic and open groups. Robotic RAMPS expanded the indications for minimally invasive surgeries for PDAC to a certain extent.
Collapse
Affiliation(s)
- Yuyao Song
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Wenbo Zou
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
- Department of General Surgery, No. 924 Hospital of PLA Joint Logistic Support Force, Guilin, China
| | - Yuanxing Gao
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Zhiming Zhao
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Zhuzeng Yin
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Chaohui Xiao
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Qu Liu
- Organ Transplantation Department, The Third Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Rong Liu
- The Faculty of Hepatopancreatobiliary Surgery, The First Medical Center, Chinese People's Liberation Army General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
107
|
Li Y, Li Y, Song Y, Liu S. Advances in research and application of photodynamic therapy in cholangiocarcinoma (Review). Oncol Rep 2024; 51:53. [PMID: 38334150 DOI: 10.3892/or.2024.8712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a disease characterized by insidious clinical manifestations and challenging to diagnose. Patients are usually diagnosed at an advanced stage and miss the opportunity for radical surgery. Therefore, effective palliative therapy is the main treatment approach for unresectable CCA. Current common palliative treatments include biliary drainage, chemotherapy, radiotherapy, targeted therapy and immunotherapy. However, these treatments only offer limited improvement in quality of life and survival. Photodynamic therapy (PDT) is a novel local treatment method that is considered a safe tumor ablation method for numerous cancers. It has shown good efficacy in various studies of CCA and is expected to become an important treatment for CCA. In the present study, the mechanisms of PDT in the treatment of CCA were systematically explored and the progress in the research of photosensitizers was discussed. The current study focused on the various PDT protocols and their therapeutic effects in CCA, with the objective of providing a new horizon for future research and clinical applications of PDT in the treatment of CCA.
Collapse
Affiliation(s)
- Yufeng Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yuhang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| | - Yinghui Song
- Central Laboratory of Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan 410005, P.R. China
| |
Collapse
|
108
|
Huang JM, Chen SH, Chen TH. Short-Term Outcomes of Conventional Laparoscopic versus Robot-Assisted Distal Pancreatectomy for Malignancy: Evidence from US National Inpatient Sample, 2005-2018. Cancers (Basel) 2024; 16:1003. [PMID: 38473361 DOI: 10.3390/cancers16051003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND The primary treatment for pancreatic cancer is surgical resection, and laparoscopic resection offers benefits over open surgery. This study aimed to compare the short-term outcomes of robot-assisted vs. conventional laparoscopic distal pancreatectomy. METHODS Data of adults ≥ 20 years old with pancreatic cancer who underwent conventional laparoscopic or robot-assisted laparoscopic distal pancreatectomy were extracted from the United States (US) Nationwide Inpatient Sample (NIS) 2005-2018 database. Comorbidities and complications were identified through the International Classification of Diseases (ICD) codes. Short-term outcomes were compared using logistic regression and included length of hospital stay (LOS), perioperative complications, in-hospital mortality, unfavorable discharge, and total hospital costs. RESULTS A total of 886 patients were included; 27% received robot-assisted, and 73% received conventional laparoscopic surgery. The mean age of all patients was 65.3 years, and 52% were females. Multivariable analysis revealed that robot-assisted surgery was associated with a significantly reduced risk of perioperative complications (adjusted odds ratio (aOR) = 0.61, 95% confidence interval (CI): 0.45-0.83) compared to conventional laparoscopic surgery. Specifically, robot-assisted surgery was associated with a significantly decreased risk of VTE (aOR = 0.35, 95% CI: 0.14-0.83) and postoperative blood transfusion (aOR = 0.37, 95% CI: 0.23-0.61). Robot-assisted surgery was associated with a significantly shorter LOS (0.76 days shorter, 95% CI: -1.43--0.09) but greater total hospital costs (18,284 USD greater, 95% CI: 4369.03-32,200.70) than conventional laparoscopic surgery. CONCLUSIONS Despite the higher costs, robot-assisted distal pancreatectomy is associated with decreased risk of complications and shorter hospital stays than conventional laparoscopic distal pancreatectomy.
Collapse
Affiliation(s)
- Jyun-Ming Huang
- Department of Surgery, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City 404327, Taiwan
- School of Medicine, China Medical University, No. 2, Yude Rd., North Dist., Taichung City 404, Taiwan
| | - Sheng-Hsien Chen
- Department of Surgery, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City 404327, Taiwan
- School of Medicine, China Medical University, No. 2, Yude Rd., North Dist., Taichung City 404, Taiwan
| | - Te-Hung Chen
- Department of Surgery, China Medical University Hospital, No. 2, Yude Rd., North Dist., Taichung City 404327, Taiwan
- School of Medicine, China Medical University, No. 2, Yude Rd., North Dist., Taichung City 404, Taiwan
| |
Collapse
|
109
|
Li K, Xue W, Lu Z, Wang S, Zheng J, Lu K, Li M, Zong Y, Xu F, Dai J, Yang Y, Sun J. Tumor-derived exosomal ADAM17 promotes pre-metastatic niche formation by enhancing vascular permeability in colorectal cancer. J Exp Clin Cancer Res 2024; 43:59. [PMID: 38413999 PMCID: PMC10898123 DOI: 10.1186/s13046-024-02991-3] [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: 12/27/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Hematological metastasis has been recognized as a crucial factor contributing to the high rates of metastasis and mortality observed in colorectal cancer (CRC). Notably, exosomes derived from cancer cells participate in the formation of CRC pre-metastatic niches; however, the mechanisms underlying their effects are largely unknown. While our preliminary research revealed the role of exosome-derived disintegrin and metalloproteinase 17 (ADAM17) in the early stages of CRC metastasis, the role of exosomal ADAM17 in CRC hematogenous metastasis remains unclear. METHODS In the present study, we isolated and purified exosomes using ultracentrifugation and identified exosomal proteins through quantitative mass spectrometry. In vitro, co-culture assays were conducted to evaluate the impact of exosomal ADAM17 on the permeability of the blood vessel endothelium. Vascular endothelial cell resistance, the cell index, membrane protein separation, flow cytometry, and immunofluorescence were employed to investigate the mechanisms underlying exosomal ADAM17-induced vascular permeability. Additionally, a mouse model was established to elucidate the role of exosomal ADAM17 in the modulation of blood vessel permeability and pre-metastatic niche formation in vivo. RESULTS Our clinical data indicated that ADAM17 derived from the circulating exosomes of patients with CRC could serve as a blood-based biomarker for predicting metastasis. The CRC-derived exosomal ADAM17 targeted vascular endothelial cells, thus enhancing vascular permeability by influencing vascular endothelial cadherin cell membrane localization. Moreover, exosomal ADAM17 mediated the formation of a pre-metastatic niche in nude mice by inducing vascular leakage, thereby promoting CRC metastasis. Nonetheless, ADAM17 selective inhibitors effectively reduced CRC metastasis in vivo. CONCLUSIONS Our results suggest that exosomal ADAM17 plays a pivotal role in the hematogenous metastasis of CRC. Thus, this protein may serve as a valuable blood-based biomarker and potential drug target for CRC metastasis intervention.
Collapse
Affiliation(s)
- Keyu Li
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Wenhua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, P.R. China
| | - Zhihua Lu
- Department of Radiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, Suzhou Dushu Lake Hospital, Suzhou, 215123, Jiangsu, China
| | - Suo Wang
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Jiayao Zheng
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Kuangyi Lu
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Ming Li
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Yang Zong
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Feng Xu
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Jiamin Dai
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China
| | - Yang Yang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu, China.
| | - Jinbing Sun
- Department of General Surgery, Changshu No. 1 People's Hospital, Affiliated Changshu Hospital of Soochow University, No. 1 Shuyuan Street, Changshu, 215500, Jiangsu, China.
| |
Collapse
|
110
|
Yu Y, Changyong E, Lin C, Wang L, Jiang T. Safety and learning curve analysis of robotic-assisted pancreaticoduodenectomy: experience of a single surgeon. J Robot Surg 2024; 18:92. [PMID: 38400999 DOI: 10.1007/s11701-024-01844-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 01/21/2024] [Indexed: 02/26/2024]
Abstract
Although prior studies have discussed learning curves (LC) of robotic-assisted pancreaticoduodenectomy (RPD), a recognized definition is lacking. This study analyzed the clinical outcomes of 85 consecutive RPD cases performed by a single surgeon to evaluate the safety and learning curve of RPD using the da Vinci Xi robotic system. There were 51 male and 34 female patients, with a median age of 64 (20-87) years. The average preoperative body weight and BMI were 64.15 ± 11.43 kg and 23.36 ± 3.33 kg/m2, respectively. The clinical outcomes of each patient were analyzed using the textbook outcome(TO), and the learning curve of the RPD was evaluated by calculating the TO rate of patients using the cumulative sum analysis method (CUSUM).The operation time (OT) was 288.92 ± 44.41 min, and the postoperative hospital stay was 10 (1-134) days. In total, 23.52% (20/85), 5.88% (5/85), 2.35% (2/85), and 5.9% (5/85) experienced grade IIIa, IIIb, IV, and V complications. A total of 46 patients achieved TO outcomes (TO group), while 39 did not (non-TO group). The smoking rate in the TO group was lower (P < 0.05) and the albumin level was higher (P < 0.05) than that in the non-TO group. The TO rate became positive after the 56th case, all patients were divided into a learning improvement group (56 cases) and a proficient group (29 cases). The total bilirubin level in the learning improvement group was lower (P < 0.05) and the bleeding volume was higher (P < 0.05).RPD is safe and effective for carefully selected patients. The learning curve was completed after 56 patients.
Collapse
Affiliation(s)
- Yang Yu
- Hapatobiliary and Pancreatic Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - E Changyong
- Hapatobiliary and Pancreatic Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Chao Lin
- Hapatobiliary and Pancreatic Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Lun Wang
- Hapatobiliary and Pancreatic Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Tao Jiang
- Hapatobiliary and Pancreatic Surgery Department, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
111
|
Zhao ZH, Huang Y, Jiang C, Lv GY, Wang M. Comparative prognosis and risk assessment in gallbladder neuroendocrine neoplasms versus adenocarcinomas. Front Endocrinol (Lausanne) 2024; 15:1326112. [PMID: 38390209 PMCID: PMC10882707 DOI: 10.3389/fendo.2024.1326112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
Background Gallbladder neuroendocrine neoplasms (GB-NENs) are a rare malignant disease, with most cases diagnosed at advanced stages, often resulting in poor prognosis. However, studies regarding the prognosis of this condition and its comparison with gallbladder adenocarcinomas (GB-ADCs) have yet to yield convincing conclusions. Methods We extracted cases of GB-NENs and GB-ADCs from the Surveillance, Epidemiology, and End Results (SEER) database in the United States. Firstly, we corrected differences in clinical characteristics between the two groups using propensity score matching (PSM). Subsequently, we visualized and compared the survival outcomes of the two groups using the Kaplan-Meier method. Next, we employed the least absolute shrinkage and selection operator (LASSO) regression and Cox regression to identify prognostic factors for GB-NENs and constructed two nomograms for predicting prognosis. These nomograms were validated with an internal validation dataset from the SEER database and an external validation dataset from a hospital. Finally, we categorized patients into high-risk and low-risk groups based on their overall survival (OS) scores. Results A total of 7,105 patients were enrolled in the study, comprising 287 GB-NENs patients and, 6,818 GB-ADCs patients. There were substantial differences in clinical characteristics between patients, and GB-NENs exhibited a significantly better prognosis. Even after balancing these differences using PSM, the superior prognosis of GB-NENs remained evident. Independent prognostic factors selected through LASSO and Cox regression were age, histology type, first primary malignancy, tumor size, and surgery. Two nomograms for prognosis were developed based on these factors, and their performance was verified from three perspectives: discrimination, calibration, and clinical applicability using training, internal validation, and external validation datasets, all of which exhibited excellent validation results. Using a cutoff value of 166.5 for the OS nomogram score, patient mortality risk can be identified effectively. Conclusion Patients with GB-NENs have a better overall prognosis compared to those with GB-ADCs. Nomograms for GB-NENs prognosis have been effectively established and validated, making them a valuable tool for assessing the risk of mortality in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
112
|
Liu R, Abu Hilal M, Besselink MG, Hackert T, Palanivelu C, Zhao Y, He J, Boggi U, Jang JY, Panaro F, Goh BKP, Efanov M, Nagakawa Y, Kim HJ, Yin X, Zhao Z, Shyr YM, Iyer S, Kakiashvili E, Han HS, Lee JH, Croner R, Wang SE, Marino MV, Prasad A, Wang W, He S, Yang K, Liu Q, Wang Z, Li M, Xu S, Wei K, Deng Z, Jia Y, van Ramshorst TME. International consensus guidelines on robotic pancreatic surgery in 2023. Hepatobiliary Surg Nutr 2024; 13:89-104. [PMID: 38322212 PMCID: PMC10839730 DOI: 10.21037/hbsn-23-132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/10/2023] [Indexed: 02/08/2024]
Abstract
Background With the rapid development of robotic surgery, especially for the abdominal surgery, robotic pancreatic surgery (RPS) has been applied increasingly around the world. However, evidence-based guidelines regarding its application, safety, and efficacy are still lacking. To harvest robust evidence and comprehensive clinical practice, this study aims to develop international guidelines on the use of RPS. Methods World Health Organization (WHO) Handbook for Guideline Development, GRADE Grid method, Delphi vote, and the AGREE-II instrument were used to establish the Guideline Steering Group, Guideline Development Group, and Guideline Secretary Group, formulate 19 clinical questions, develop the recommendations, and draft the guidelines. Three online meetings were held on 04/12/2020, 30/11/2021, and 25/01/2022 to vote on the recommendations and get advice and suggestions from all involved experts. All the experts focusing on minimally invasive surgery from America, Europe and Oceania made great contributions to this consensus guideline. Results After a systematic literature review 176 studies were included, 19 questions were addressed and 14 recommendations were developed through the expert assessment and comprehensive judgment of the quality and credibility of the evidence. Conclusions The international RPS guidelines can guide current practice for surgeons, patients, medical societies, hospital administrators, and related social communities. Further randomized trials are required to determine the added value of RPS as compared to open and laparoscopic surgery.
Collapse
Affiliation(s)
- Rong Liu
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Mohammed Abu Hilal
- Department of Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Marc G. Besselink
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Thilo Hackert
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Chinnusamy Palanivelu
- Department of Minimal Invasive Hernia Surgery, GEM Hospital and Research Centre, Chennai, Tamil Nadu, India
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital Beijing, Beijing, China
| | - Jin He
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ugo Boggi
- Division of General and Transplant Surgery, University of Pisa, Pisa, Italy
| | - Jin-Young Jang
- Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Fabrizio Panaro
- Department of Surgery/Division of HBP Surgery & Transplantation, University of Montpellier, Montpellier, France
| | - Brian K. P. Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore
| | - Mikhail Efanov
- Department of Hepato-Pancreato-Biliary Surgery, Moscow Clinical Scientific Center, Moscow, Russia
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, Shinjuku, Tokyo, Japan
| | - Hong-Jin Kim
- Department of Surgery, Yeungnam University College of Medicine, Daegu, Korea
| | - Xiaoyu Yin
- Department of Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiming Zhao
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yi-Ming Shyr
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei
| | - Shridhar Iyer
- Division of Hepatobiliary, Pancreatic Surgery and Liver Transplantation, National University Hospital, Singapore, Singapore
| | - Eli Kakiashvili
- Department of Surgery, Galilee Medical Center, Nahariya, Israel
| | - Ho-Seong Han
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Korea
| | - Jae Hoon Lee
- Division of Hepato-Biliary and Pancreatic Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Roland Croner
- Department of General-, Vascular-, Visceral- and Transplant Surgery, University Hospital Magdeburg, Magdeburg, Germany
| | - Shin-E Wang
- Division of General Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei
| | - Marco Vito Marino
- General Surgery Department, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy
| | - Arun Prasad
- Department of General and Minimal Access Surgery and Robotic Surgery, Indraprastha Apollo Hospitals, New Delhi, India
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kehu Yang
- EvidenceBased Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Qu Liu
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zizheng Wang
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Mengyang Li
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Shuai Xu
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Kongyuan Wei
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Zhaoda Deng
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yuze Jia
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Tess M. E. van Ramshorst
- Department of Surgery, Poliambulanza Foundation Hospital, Brescia, Italy
- Amsterdam UMC, location University of Amsterdam, Department of Surgery, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
113
|
Almalki WH. NEAT1 in inflammatory infectious diseases: An integrated perspective on molecular modulation. Pathol Res Pract 2024; 254:154956. [PMID: 38218038 DOI: 10.1016/j.prp.2023.154956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
The long non-coding RNA (lncRNA), NEAT1, has emerged as a central figure in the intricate network of molecular regulators in inflammatory infectious diseases (IIDs). The review initiates a comprehensive exploration of NEAT1's multifaceted roles and molecular interactions in the context of these complex diseases. The study begins by acknowledging the global health burden of IIDs, underscoring the urgency for innovative insights into their pathogenesis and therapeutic avenues. NEAT1 is introduced as a pivotal lncRNA with growing relevance in immune responses and inflammatory processes. The core of this review unravels the NEAT1 landscape, elucidating its involvement in the modulation of immune signalling pathways, regulation of inflammatory cytokines, and interactions with various immune cells during infection. It explores NEAT1's role in orchestrating immune responses and balancing host defence mechanisms with the risk of immunopathology. Furthermore, the review underscores the clinical significance of NEAT1 in infectious diseases, discussing its associations with disease severity, prognosis, and potential as a diagnostic and therapeutic target. It provides insights into ongoing research endeavours aimed at harnessing NEAT1 for innovative disease management strategies, including developing RNA-based therapeutics. Concluding on a forward-looking note, the review highlights the broader implications of NEAT1 in the context of emerging infectious diseases and the possibility for precision medicine approaches that leverage NEAT1's regulatory capacities. In summary, this review illuminates the pivotal role of NEAT1 in IIDs by navigating its complex landscape, offering profound insights into its implications for disease pathogenesis and the development of targeted therapies.
Collapse
Affiliation(s)
- Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| |
Collapse
|
114
|
Mohanan A, Biju P, V B, V G. Unraveling Proto-Oncogene (ErbB2) Expression in Patients With Carcinoma Head of Pancreas and Chronic Pancreatitis Patients: A Case-Control Study. Cureus 2024; 16:e54859. [PMID: 38533139 PMCID: PMC10964396 DOI: 10.7759/cureus.54859] [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] [Accepted: 02/25/2024] [Indexed: 03/28/2024] Open
Abstract
Background The pre-malignant tendency of the normal, non-affected portion of the pancreas is not as well explored as the multicentricity documented in pancreatic cancer cases. In order to ascertain the expression of inflammatory markers and Erythroblastic Oncogene B (ErbB2) in the non-affected pancreas in patients with pancreatic cancer, a case-control study was carried out. Materials and methods In patients who underwent pancreatoduodenectomy for pancreatic cancer (PC), pro-inflammatory genes and a tumor marker, erythroblastic oncogene 2 (ErbB2) in the epidermal growth factor receptor family were analyzed in the pancreatic tissue at the cut surface of the normal pancreas using qRT-PCR. Twenty patients diagnosed with Chronic pancreatitis (CP) after Frey's surgical procedure were selected, and their pancreatic tissues were analyzed as controls. The HPLC-purified primers were designed using National Center for Biotechnology Information (NCBI) software. The primer's specificity was verified for gene expression analysis using the Basic Local Alignment Search Tool (BLAST). The genes under study were normalized using β-actin as the housekeeping gene, and the 2-ddct method was used to compute the fold change compared to the control sample. Results Patients with margin-positive were not included. Pro-inflammatory genes (TNF-α, NF-kβ, and COX-2) had significantly lower foldchange in PC patients compared to the CP group. The CP control group had higher levels of IL-6 gene expression than the PC group. Patients with pancreatic cancer had a considerably higher expression of the ErbB2 gene than patients with CP. Conclusion The upregulated ErbB2 gene in the unaffected pancreatic tissue of pancreatic cancer patients, when compared to controls, indicates that the remaining pancreas may have the capacity to cause cancer. Proto-oncogene may play a role in the pathophysiologic process in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Abhina Mohanan
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Pottakkat Biju
- Surgical Gastroenterology, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Balasubramaniyan V
- Biochemistry, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| | - Gladwin V
- Anatomy, Jawaharlal Institute of Postgraduate Medical Education & Research, Puducherry, IND
| |
Collapse
|
115
|
Liu B, Wu T, Lin B, Liu X, Liu Y, Song G, Fan C, Ouyang G. Periostin-TGF-β feedforward loop contributes to tumour-stroma crosstalk in liver metastatic outgrowth of colorectal cancer. Br J Cancer 2024; 130:358-368. [PMID: 38097742 PMCID: PMC10844286 DOI: 10.1038/s41416-023-02516-3] [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: 02/24/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND This study aimed to investigate the underlying mechanisms of matricellular protein periostin (POSTN) on tumour-stroma crosstalk in the liver metastatic microenvironment of colorectal cancer (CRC). METHODS Postn-knockout mice and hepatic Postn-overexpressing mice were used to investigate the functions of POSTN on the formation of fibrotic microenvironment and the tumour-stroma crosstalk in the liver metastatic microenvironment of CRC. Clinical samples and database were analyzed to show the correlation between POSTN expression and fibrotic features and TGF-β signalling in metastatic livers of CRC. RESULTS POSTN deficiency reduced hepatic stellate cell (HSC) activation and liver metastasis, whereas POSTN overexpression in the liver significantly augmented the formation of a fibrotic microenvironment to support the liver metastatic growth of CRC cells in mice. Moreover, HSC-derived POSTN promoted TGF-β1 expression in CRC cells through the integrin/FAK/ERK/STAT3 pathway; conversely, tumour cell-derived TGF-β1 induced POSTN expression in HSCs via the Smad pathway. POSTN levels correlated with fibrotic features and TGF-β signalling in metastatic liver tissues of CRC patients. CONCLUSIONS POSTN and TGF-β1 cooperatively contribute to the tumour-stroma crosstalk by forming a supporting fibrotic microenvironment to promote liver metastasis of CRC cells via the POSTN/integrin/FAK/ERK/STAT3/TGF-β axis in tumour cells and TGF-β/Smad/POSTN signalling in activated HSCs.
Collapse
Affiliation(s)
- Bin Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Tiantian Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Biyu Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Xingxing Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
| | - Yingfu Liu
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China
| | - Gang Song
- Department of Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, China.
| | - Chuannan Fan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Gaoliang Ouyang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
- Department of Hematology, the First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
116
|
Zhou X, Li D, Xia S, Ma X, Li R, Mu Y, Liu Z, Zhang L, Zhou Q, Zhuo W, Ding K, Lin A, Liu W, Liu X, Zhou T. RNA-based modulation of macrophage-mediated efferocytosis potentiates antitumor immunity in colorectal cancer. J Control Release 2024; 366:128-141. [PMID: 38104775 DOI: 10.1016/j.jconrel.2023.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/28/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023]
Abstract
Tumor-associated macrophages play pivotal roles in tumor progression and metastasis. Macrophage-mediated clearance of apoptotic cells (efferocytosis) supports inflammation resolution, contributing to immune evasion in colorectal cancers. To reverse this immunosuppressive process, we propose a readily translatable RNA therapy to selectively inhibit macrophage-mediated efferocytosis in tumor microenvironment. A clinically approved lipid nanoparticle platform (LNP) is employed to encapsulate siRNA for the phagocytic receptor MerTK (siMerTK), enabling selective MerTK inhibition in the diseased organ. Decreased MerTK expression in tumor-associated macrophages results in apoptotic cell accumulation and immune activation in tumor microenvironment, leading to suppressed tumor growth and better survival in both liver and peritoneal metastasis models of colorectal cancers. siMerTK delivery combined with PD-1 blockade further produces enhanced antimetastatic efficacy with reactivated intratumoral immune milieu. Collectively, LNP-based siMerTK delivery combined with immune checkpoint therapy may present a feasible modality for metastatic colorectal cancer therapy.
Collapse
Affiliation(s)
- Xuefei Zhou
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China.
| | - Dezhi Li
- Department of Oncology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Shenglong Xia
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Cancer Center, Zhejiang University, Hangzhou 310058, China
| | - Xixi Ma
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China
| | - Rong Li
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yongli Mu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zimo Liu
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lu Zhang
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Quan Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wei Zhuo
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Kefeng Ding
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wei Liu
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu 322000, China
| | - Xiangrui Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China; Cancer Center, Zhejiang University, Hangzhou 310058, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China.
| | - Tianhua Zhou
- Cancer Center, Zhejiang University, Hangzhou 310058, China; Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang 310020, China.
| |
Collapse
|
117
|
Zhang Y, Hao M, Yang X, Zhang S, Han J, Wang Z, Chen HN. Reactive oxygen species in colorectal cancer adjuvant therapies. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166922. [PMID: 37898425 DOI: 10.1016/j.bbadis.2023.166922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/27/2023] [Accepted: 10/18/2023] [Indexed: 10/30/2023]
Abstract
Colorectal cancer (CRC), a prevalent global malignancy, often necessitates adjuvant therapies such as chemotherapy, radiotherapy, targeted therapy, and immunotherapy to mitigate tumor burden in advanced stages. The efficacy of these therapies is significantly influenced by reactive oxygen species (ROS). Previous research underscores the pivotal role of ROS in gut pathology, targeted therapy, and drug resistance. ROS-mediated CRC adjuvant therapies encompass a myriad of mechanisms, including cell death and proliferation, survival and cell cycle, DNA damage, metabolic reprogramming, and angiogenesis. Preliminary clinical trials have begun to unveil the potential of ROS-manipulating therapy in enhancing CRC adjuvant therapies. This review aims to provide a comprehensive synthesis of studies exploring the role of ROS in CRC adjuvant therapies.
Collapse
Affiliation(s)
- Yang Zhang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mengqiu Hao
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xuyang Yang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Su Zhang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziqiang Wang
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hai-Ning Chen
- Colorectal Cancer Center and Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China; Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, Frontiers Science Center for Disease-related Molecular Network and National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
118
|
Liu J, Xu H, Gao S, Liu L, Qu J, Ohulchanskyy TY. Combining near infrared fluorescence and laser speckle imaging with optical tissue clearing for in vivo transcranial monitoring of cerebral blood vessels damaged by photodynamic nanoformulation. BIOMEDICAL OPTICS EXPRESS 2024; 15:924-937. [PMID: 38404313 PMCID: PMC10890862 DOI: 10.1364/boe.513820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/27/2024]
Abstract
In vivo near infrared (NIR) fluorescence imaging and laser speckle contrast imaging (LSCI) are emerging optical bioimaging modalities, which can provide information on blood vessels morphology, volume and the blood flow velocity. Optical tissue clearing (OTC) technique addresses a light scattering problem in optical bioimaging, which is imperative for the transcranial brain imaging. Herein, we report an approach combining NIR fluorescence and LSC microscopy imaging with OTC. A liposomal nanoformulation comprising NIR fluorescent dye ICG and photosensitizer BPD was synthesized and injected intravenously into mouse with OTC treated skull. Transcranial excitation of BPD in nanoliposomes resulted in the localized, irradiation dose dependent photodynamic damage of the brain blood vessels, which was manifested both in NIR fluorescence and LSC transcranial imaging, revealing changes in the vessels morphology, volume and the blood flow rate. The developed approach allows for bimodal imaging guided, localized vascular PDT of cancer and other diseases.
Collapse
Affiliation(s)
- Jiantao Liu
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Hao Xu
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Siqi Gao
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
- Engineering Research Center of Optical
Instrument and System, Ministry of Education, Shanghai Key Lab of
Modern Optical System, School of Optical-Electrical and Computer
Engineering, University of Shanghai for Science and
Technology, Shanghai, China
| | - Tymish Y. Ohulchanskyy
- Key Laboratory of Optoelectronic Devices
and Systems of Ministry of Education and Guangdong Province, College
of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
119
|
Gao F, Xie Q, Zhao X, Yang M, Jiang K, Zhang L, Mao T, Wu H. Preliminary exploration of hepatic parenchymal near-infrared fluorescence imaging technique via retrograde biliary approach: a feasibility study (with video). Sci Rep 2024; 14:2380. [PMID: 38286815 PMCID: PMC10824724 DOI: 10.1038/s41598-024-52904-9] [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: 10/16/2023] [Accepted: 01/24/2024] [Indexed: 01/31/2024] Open
Abstract
This paper explores the feasibility and principle of hepatic parenteral fluorescence imaging technology after retrograde injection of indocyanine green (ICG) through endoscopic nasobiliary drainage (ENBD). The data were collected from 53 patients with cholecystolithiasis and choledocholithiasis, from October 2022 to March 2023, diagnosed by fluorescence imaging technique retrograde biliary approach (FIT-RB). We divided the patients into two groups according to the features of liver parenchyma, the poor group (n = 34, including scattered or no imaging) and the good group (n = 19, regular uniform imaging). We compared and analyzed the perioperative results of the two groups and explored the influencing factors of the success of FIT-RB and the ICG concentration suitable for this imaging technique. The good imaging rate of the 53 enrolled cases was 35.8%. The bilirubin level before ENBD and laparoscopic cholecystectomy in the poor group was significantly higher than that in the good group (P < 0.001). The proportion of higher ICG concentrations (0.5 mg/mL) was significantly higher in the good group (P = 0.028). Our results demonstrated that the success rate of good imaging was 4.53 times higher than that of low-dose ICG (0.125 or 0.25 mg/L) cases at 0.5 mg/ml of ICG. The level of total bilirubin and direct bilirubin were negatively correlated with the imaging effect, and total bilirubin and direct bilirubin levels were important predictors of the efficacy of FIT-RB. FIT-RB is safe and feasible in patients with low site bilirubin levels. An ICG concentration of 0.5 mg/ml may be ideal for implementing this technique.
Collapse
Affiliation(s)
- Fengwei Gao
- Liver Transplantation Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Qingyun Xie
- Liver Transplantation Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan Province, China
| | - Xin Zhao
- Department of Hepato-Pancreato-Biliary Surgery, The People's Hospital of Leshan, Leshan, 614000, China
| | - Manyu Yang
- North Sichuan Medical College, Nanchong, 637000, China
| | - Kangyi Jiang
- Department of Hepato-Pancreato-Biliary Surgery, The People's Hospital of Leshan, Leshan, 614000, China
| | - Ling Zhang
- Department of Hepato-Pancreato-Biliary Surgery, The People's Hospital of Leshan, Leshan, 614000, China
| | - Tianyang Mao
- North Sichuan Medical College, Nanchong, 637000, China
| | - Hong Wu
- Liver Transplantation Center, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, No. 37, Guoxue Lane, Wuhou District, Chengdu, 610041, Sichuan Province, China.
| |
Collapse
|
120
|
Underwood PW, Ruff SM, Pawlik TM. Update on Targeted Therapy and Immunotherapy for Metastatic Colorectal Cancer. Cells 2024; 13:245. [PMID: 38334637 PMCID: PMC10854977 DOI: 10.3390/cells13030245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/21/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Metastatic colorectal cancer remains a deadly malignancy and is the third leading cause of cancer-related death. The mainstay of treatment for metastatic colorectal cancer is chemotherapy, but unfortunately, even with recent progress, overall survival is still poor. Colorectal cancer is a heterogeneous disease, and the underlying genetic differences among tumors can define the behavior and prognosis of the disease. Given the limitations of cytotoxic chemotherapy, research has focused on developing targeted therapy based on molecular subtyping. Since the early 2000s, multiple targeted therapies have demonstrated efficacy in treating metastatic colorectal cancer and have received FDA approval. The epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and DNA mismatch repair pathways have demonstrated promising results for targeted therapies. As new gene mutations and proteins involved in the oncogenesis of metastatic colorectal cancer are identified, new targets will continue to emerge. We herein provide a summary of the updated literature regarding targeted therapies for patients with mCRC.
Collapse
Affiliation(s)
| | | | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, 395 W. 12th Ave., Suite 670, Columbus, OH 43210, USA; (P.W.U.); (S.M.R.)
| |
Collapse
|
121
|
Ciardiello D, Petrillo A. Editorial: Optimizing the first-line treatment for metastatic colorectal cancer. Front Oncol 2024; 14:1366383. [PMID: 38327740 PMCID: PMC10847841 DOI: 10.3389/fonc.2024.1366383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024] Open
Affiliation(s)
- Davide Ciardiello
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IEO, IRCCS, Milan, Italy
| | | |
Collapse
|
122
|
Zhang C, Zhang Y, Dong Y, Zi R, Wang Y, Chen Y, Liu C, Wang J, Wang X, Li J, Liang H, Ou J. Non-alcoholic fatty liver disease promotes liver metastasis of colorectal cancer via fatty acid synthase dependent EGFR palmitoylation. Cell Death Discov 2024; 10:41. [PMID: 38263401 PMCID: PMC10805926 DOI: 10.1038/s41420-023-01770-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/02/2023] [Accepted: 12/07/2023] [Indexed: 01/25/2024] Open
Abstract
Liver metastasis is the major reason for most of colorectal cancer (CRC) related deaths. Accumulating evidence indicates that CRC patients with non-alcoholic fatty liver disease (NAFLD) are at a greater risk of developing liver metastasis. With the growing prevalence of NAFLD, a better understanding of the molecular mechanism in NAFLD-driven CRC liver metastasis is needed. In this study, we demonstrated that NAFLD facilitated CRC liver metastasis as a metabolic disorder and promoted the stemness of metastatic CRC cells for their colonization and outgrowth in hepatic niches. Metabolically, the lipid-rich microenvironment in NAFLD activated de novo palmitate biosynthesis in metastatic CRC cells via upregulating fatty acid synthase (FASN). Moreover, increased intracellular palmitate bioavailability promoted EGFR palmitoylation to enhance its protein stability and plasma membrane localization. Furthermore, we demonstrated that the FDA-approved FASN inhibitor orlistat could reduce NAFLD-activated endogenous palmitate production, thus inhibiting palmitoylation of EGFR to suppress CRC cell stemness and restrict liver metastasis in synergy with conventional chemotherapy. These findings reveal that the NAFLD metabolic microenvironment boosts endogenous palmitate biosynthesis in metastatic CRC cells and promotes cell stemness via EGFR palmitoylation, and FASN inhibitor orlistat could be a candidate adjuvant drug to suppress liver metastasis in CRC patients with NAFLD.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yue Zhang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yan Dong
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Ruiyang Zi
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yijie Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yanrong Chen
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chengxiang Liu
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Junyi Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Xuesong Wang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Jianjun Li
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Houjie Liang
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
| | - Juanjuan Ou
- Department of Oncology and Southwest Cancer Centre, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China.
- Jinfeng Laboratory, 401329, Chongqing, China.
| |
Collapse
|
123
|
Li CMY, Tomita Y, Dhakal B, Tin T, Li R, Wright JA, Vrbanac L, Woods SL, Drew P, Price T, Smith E, Maddern GJ, Fenix K. Generation and assessment of cytokine-induced killer cells for the treatment of colorectal cancer liver metastases. Cancer Immunol Immunother 2024; 73:6. [PMID: 38231291 PMCID: PMC10794456 DOI: 10.1007/s00262-023-03591-4] [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: 09/20/2023] [Accepted: 11/04/2023] [Indexed: 01/18/2024]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide. Cytokine-induced killer (CIK) cells are an adoptive immunotherapy reported to have strong anti-tumour activity across a range of cancers. They are a heterogeneous mix of lymphoid cells generated by culturing human peripheral blood mononuclear cells with cytokines and monoclonal antibodies in vitro. In this study, we investigated the yield and function of CIK cells generated from patients with CRC liver metastases. We first showed that CIK cells generated in serum free medium X-VIVO 15 were comparable to those from RPMI medium with 10% FBS in terms of the number and percentages of the main subsets of cells in the CIK culture, and the intracellular levels of granzyme B and perforin, and the pro-inflammatory cytokines IL-2, IFN-γ and TNF-α. The CIK cells were cytotoxic to CRC cell lines grown in 2D cultures or as spheroids, and against autologous patient-derived tumour organoids. Donor attributes such as age, sex, or prior chemotherapy exposure had no significant impact on CIK cell numbers or function. These results suggest that functional CIK cells can be generated from patients with CRC liver metastatic disease, and support further investigations into the therapeutic application of autologous CIK cells in the management of patients with CRC liver metastases.
Collapse
Affiliation(s)
- Celine Man Ying Li
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
| | - Yoko Tomita
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
- Medical Oncology, The Queen Elizabeth Hospital, The University of Adelaide, Adelaide, 5011, Australia
| | - Bimala Dhakal
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
| | - Teresa Tin
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
| | - Runhao Li
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
- Medical Oncology, The Queen Elizabeth Hospital, The University of Adelaide, Adelaide, 5011, Australia
| | - Josephine A Wright
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, 5005, Australia
| | - Laura Vrbanac
- Department of Medical Specialties, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
| | - Susan L Woods
- Precision Medicine, South Australian Health and Medical Research Institute, Adelaide, 5005, Australia
- Department of Medical Specialties, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
| | - Paul Drew
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
| | - Timothy Price
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
- Medical Oncology, The Queen Elizabeth Hospital, The University of Adelaide, Adelaide, 5011, Australia
| | - Eric Smith
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
- Medical Oncology, The Queen Elizabeth Hospital, The University of Adelaide, Adelaide, 5011, Australia
| | - Guy J Maddern
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia
| | - Kevin Fenix
- Discipline of Surgery, Adelaide Medical School, The University of Adelaide, Adelaide, 5005, Australia.
- The Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Adelaide, 5011, Australia.
| |
Collapse
|
124
|
Hajibandeh S, Scarpa E, Kaur N, Alessandri G, Kumar N. Prognostic significance of socioeconomic deprivation in patients with colorectal liver metastasis undergoing liver resection: a retrospective cohort study. Langenbecks Arch Surg 2024; 409:31. [PMID: 38191745 DOI: 10.1007/s00423-023-03220-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/29/2023] [Indexed: 01/10/2024]
Abstract
AIMS To evaluate the effect of socioeconomic deprivation on overall survival (OS) in patients undergoing liver resection for colorectal liver metastasis (CRLM). METHODS The STROCSS guideline for observational studies was followed to conduct a single-centre retrospective cohort study. All consecutive patients undergoing resection of CRLM between 2013 and 2021 were considered eligible for inclusion. The Welsh Index of Multiple Deprivation (WIMD) rank was used to determine socioeconomic deprivation status of each patient. Prognostic significance of socioeconomic deprivation was determined by Kaplan-Meier survival statistics and stepwise Cox proportional-hazards regression model. RESULTS A total of 455 patients were eligible for inclusion; 237 patients were classed as least socioeconomically deprived and 218 patients as most socioeconomically deprived. Kaplan-Meier survival statistics showed that socioeconomic deprivation was associated with significantly lower probability of overall survival (HR: 1.55, 95% CI 1.23-1.95; logrank test: P = 0.0001). The stepwise Cox proportional-hazards regression analysis identified socioeconomic deprivation as predictor of OS (HR: 1.56, P = 0.0003) alongside the following variables: ASA status 1 (HR: 0.43, P = 0.0349), presence of extrahepatic disease (HR: 1.51, P = 0.0075), number of tumours (HR: 1.07, P = 0.0221), size of largest tumour (HR: 1.01, P = 0.0003), extended hemihepatectomy (HR: 3.24, P = 0.0018) and absence of recurrence (HR: 0.55, P < 0.0001). CONCLUSIONS Socioeconomic deprivation reduces the probability of long-term overall survival following liver resection in patients with CRLM. This should be taken into account at different levels of health care planning for management of patients with CRLM including preoperative risk assessment, health care need assessment and allocation of resources.
Collapse
Affiliation(s)
- Shahab Hajibandeh
- Cardiff Liver Unit, University Hospital of Wales, Cardiff & Vale NHS Trust, Cardiff, CF14 4XW, UK.
| | - Emanuele Scarpa
- Cardiff Liver Unit, University Hospital of Wales, Cardiff & Vale NHS Trust, Cardiff, CF14 4XW, UK
| | - Namratha Kaur
- Cardiff Liver Unit, University Hospital of Wales, Cardiff & Vale NHS Trust, Cardiff, CF14 4XW, UK
| | - Giorgio Alessandri
- Cardiff Liver Unit, University Hospital of Wales, Cardiff & Vale NHS Trust, Cardiff, CF14 4XW, UK
| | - Nagappan Kumar
- Cardiff Liver Unit, University Hospital of Wales, Cardiff & Vale NHS Trust, Cardiff, CF14 4XW, UK
| |
Collapse
|
125
|
Zhang D, Zhang M, Pang Y, Li M, Ma W. Folic Acid-Modified Long-Circulating Liposomes Loaded with Sulfasalazine For Targeted Induction of Ferroptosis in Melanoma. ACS Biomater Sci Eng 2024; 10:588-598. [PMID: 38117929 DOI: 10.1021/acsbiomaterials.3c01223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Melanoma is a malignant tumor that originates from melanocytes. The incidence of melanoma is increasing worldwide, partially because of its insensitivity to radiotherapy or chemotherapy. Therefore, effective treatments for melanoma are urgently required. In this study, we employed folic acid-modified sulfasalazine long-circulating liposomes (FA-SSZ-Lips) to precisely target drug delivery to melanoma cells, eliciting ferroptosis effectively. The synthesized FA-SSZ-Lips were characterized as small spheres of a double-layer membrane, a particle size of 110.1 nm, and a ζ-potential of -22.8 ± 0.66 mV. FA-SSZ-Lips are effective drug carriers with SSZ-loading ratio and SSZ release rate of 6.2 ± 0.10%, and 72.63 ± 1.40%, respectively. The liposomes enhanced SSZ solubility, and the folic acid modifications increased the liposome targeting to melanoma cells. Compared with SSZ alone, FA-SSZ-Lips more strongly inhibited B16F10 cell growth, significantly disrupted the intracellular redox balance, and induced ferroptosis. After treatment, considerable differences were observed in the tumor volumes between FA-SSZ-Lips and phosphate-buffered saline control groups. The tumor growth-inhibition value of the FA-SSZ-Lips group reached 70.09%. Thus, FA-SSZ-Lips exhibited favorable antitumor effects in vitro and in vivo and are a promising strategy for melanoma treatment.
Collapse
Affiliation(s)
- Dong Zhang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Mogen Zhang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Yunyan Pang
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Meiling Li
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| | - Weiyuan Ma
- Department of Dermatology, Affiliated Hospital of Weifang Medical University, Weifang 261031, China
| |
Collapse
|
126
|
Coffman-D'Annibale K, Myojin Y, Monge C, Xie C, Hrones DM, Wood BJ, Levy EB, Kleiner D, Figg WD, Steinberg SM, Redd B, Greten TF. VB-111 (ofranergene obadenovec) in combination with nivolumab in patients with microsatellite stable colorectal liver metastases: a single center, single arm, phase II trial. J Immunother Cancer 2024; 12:e008079. [PMID: 38184304 PMCID: PMC10773432 DOI: 10.1136/jitc-2023-008079] [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] [Accepted: 12/13/2023] [Indexed: 01/08/2024] Open
Abstract
BACKGROUND Microsatellite stable colorectal liver metastases (MSS CLM) maintain an immunosuppressive tumor microenvironment (TME). Historically, immune-based approaches have been ineffective. VB-111 (ofranergene obadenovec) is a genetically-modified adenoviral vector targeting the TME; its unique dual mechanism induces an immune response and disrupts neovascularization. Checkpoint inhibition may synergize the immune response induced by viral-mediated anti-angiogenic gene therapy. We aimed to examine the safety and antitumor activity of VB-111 and nivolumab in patients with refractory MSS CLM and to characterize immunological treatment-response. METHODS This was a phase II study of adult patients with histologically-confirmed MSS CLM who progressed on prior therapy. A priming dose of VB-111 1×1013 viral particles was given intravenously 2 weeks prior to starting biweekly nivolumab 240 mg and continued every 6 weeks. The combination continued until disease progression or unacceptable toxicity. The primary objectives were overall response rate and safety/tolerability. Secondary objectives included median overall survival and progression-free survival. Correlative studies were performed on paired tumor biopsies and blood. RESULTS Between August 2020 and December 2021, 14 patients were enrolled with median age 50.5 years (40-75), and 14% were women. Median follow-up was 5.5 months. Of the 10 evaluable patients, the combination of VB-111 and nivolumab failed to demonstrate radiographic responses; at best, 2 patients had stable disease. Median overall survival was 5.5 months (95% CI: 2.3 to 10.8), and median progression-free survival was 1.8 months (95% CI: 1.4 to 1.9). The most common grade 3-4 treatment-related adverse events were fever/chills, influenza-like symptoms, and lymphopenia. No treatment-related deaths were reported. Qualitative analysis of immunohistochemical staining of paired tumor biopsies did not demonstrate significant immune infiltration after treatment, except for one patient who had exceptional survival (26.0 months). Immune analysis of peripheral blood mononuclear cells showed an increase of PD-1highKi67highCD8+ T cells and HLA-DRhigh T cells after VB-111 priming dose. Plasma cytokines interleukin-10 and tumor necrosis factor-α increased after treatment with both drugs. CONCLUSION In patients with MSS CLM, VB-111 and nivolumab did not improve overall response rate or survival but were tolerated with minimal toxicities. While challenging to distinguish between antiviral or antitumor, correlative studies demonstrated an immune response with activation and proliferation of CD8+ T cells systemically that was poorly sustained. TRIAL REGISTRATION NUMBER NCT04166383.
Collapse
Affiliation(s)
- Kelley Coffman-D'Annibale
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuta Myojin
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Cecilia Monge
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Changqing Xie
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Donna Mabry Hrones
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Bradford J Wood
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center & Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Elliot B Levy
- Center for Interventional Oncology, Radiology and Imaging Sciences, NIH Clinical Center & Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - David Kleiner
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - William Douglas Figg
- Molecular Pharmacology Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Seth M Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Bernadette Redd
- Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tim F Greten
- Gastrointestinal Malignancies Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
- Liver Cancer Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
127
|
Yang J, Huang J, Han D, Ma X. Artificial Intelligence Applications in the Treatment of Colorectal Cancer: A Narrative Review. Clin Med Insights Oncol 2024; 18:11795549231220320. [PMID: 38187459 PMCID: PMC10771756 DOI: 10.1177/11795549231220320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 11/26/2023] [Indexed: 01/09/2024] Open
Abstract
Colorectal cancer is the third most prevalent cancer worldwide, and its treatment has been a demanding clinical problem. Beyond traditional surgical therapy and chemotherapy, newly revealed molecular mechanisms diversify therapeutic approaches for colorectal cancer. However, the selection of personalized treatment among multiple treatment options has become another challenge in the era of precision medicine. Artificial intelligence has recently been increasingly investigated in the treatment of colorectal cancer. This narrative review mainly discusses the applications of artificial intelligence in the treatment of colorectal cancer patients. A comprehensive literature search was conducted in MEDLINE, EMBASE, and Web of Science to identify relevant papers, resulting in 49 articles being included. The results showed that, based on different categories of data, artificial intelligence can predict treatment outcomes and essential guidance information of traditional and novel therapies, thus enabling individualized treatment strategy selection for colorectal cancer patients. Some frequently implemented machine learning algorithms and deep learning frameworks have also been employed for long-term prognosis prediction in patients with colorectal cancer. Overall, artificial intelligence shows encouraging results in treatment strategy selection and prognosis evaluation for colorectal cancer patients.
Collapse
Affiliation(s)
- Jiaqing Yang
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
- West China School of Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Huang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
| | - Deqian Han
- Department of Oncology, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Biotherapy, West China Hospital and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
128
|
Ghaffar S, Shahnoor S, Khan AM, Asif A, Fida M, Oduoye MO, Nafula WP. CRP Albumin Ratio: A novel noninvasive and cost-effective method for assessing the severity of acute pancreatitis. Health Sci Rep 2024; 7:e1801. [PMID: 38196567 PMCID: PMC10774490 DOI: 10.1002/hsr2.1801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 01/11/2024] Open
Abstract
Introduction Acute pancreatitis is a significant and potentially life-threatening gastrointestinal disorder that requires prompt and accurate diagnosis for effective treatment. Current diagnostic methods often involve expensive and inaccessible imaging studies, such as CT scans, limiting their utility in emergency settings and underserved areas. Aim This correspondence discusses an innovative and cost-effective approach to assessing the severity of acute pancreatitis, focusing on the C-Reactive Protein-Albumin Ratio (CAR). Methodology We searched relevant articles and studies from 2015 till date on PubMed, Web of Science, and Google Scholar using these keywords: "C-Reactive Protein", "Albumin Ratio", "Acute pancreatitis", "Cost-effective", "Non-invasive", and "Severity". Result The CAR diagnostic method involves a simple blood test that measures the levels of C-Reactive Protein (CRP) and albumin, both commonly used markers for assessing inflammation. Elevated CRP and decreased albumin levels are indicative of inflammation, and the CAR has shown a strong positive correlation with the severity of acute pancreatitis. This method offers a non-invasive, time-efficient, and cost-friendly alternative to traditional diagnostic techniques. Conclusion The potential of CAR as an assessment tool for the severity of acute pancreatitis is highlighted, especially in resource-limited settings. This innovation holds promise for improving the timely and accurate diagnosis of acute pancreatitis, ultimately enhancing patient outcomes and reducing mortality rates.
Collapse
Affiliation(s)
- Sania Ghaffar
- Department of Internal MedicineAyub Medical CollegeAbbottabadPakistan
| | - Syeda Shahnoor
- Department of Internal MedicineDow University of Health SciencesKarachiPakistan
| | - Abdul Moiz Khan
- Department of Internal MedicineAyub Medical CollegeAbbottabadPakistan
| | - Aimen Asif
- Department of Internal MedicineAyub Medical CollegeAbbottabadPakistan
| | - Maryam Fida
- Department of Internal MedicineAyub Medical CollegeAbbottabadPakistan
| | - Malik Olatunde Oduoye
- Department of Research at Medical Research Circle in BukavuDemocratic Republic of CongoBukavuCongo
| | - Wechuli Polyne Nafula
- Department of clinical medicineJomo Kenyatta University of Agriculture and TechnologyNairobiKenya
| |
Collapse
|
129
|
Consoli V, Fallica AN, Sorrenti V, Pittalà V, Vanella L. Novel Insights on Ferroptosis Modulation as Potential Strategy for Cancer Treatment: When Nature Kills. Antioxid Redox Signal 2024; 40:40-85. [PMID: 37132605 PMCID: PMC10824235 DOI: 10.1089/ars.2022.0179] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
Significance: The multifactorial nature of the mechanisms implicated in cancer development still represents a major issue for the success of established antitumor therapies. The discovery of ferroptosis, a novel form of programmed cell death distinct from apoptosis, along with the identification of the molecular pathways activated during its execution, has led to the uncovering of novel molecules characterized by ferroptosis-inducing properties. Recent advances: As of today, the ferroptosis-inducing properties of compounds derived from natural sources have been investigated and interesting findings have been reported both in vitro and in vivo. Critical Issues: Despite the efforts made so far, only a limited number of synthetic compounds have been identified as ferroptosis inducers, and their utilization is still limited to basic research. In this review, we analyzed the most important biochemical pathways involved in ferroptosis execution, with particular attention to the newest literature findings on canonical and non-canonical hallmarks, together with mechanisms of action of natural compounds identified as novel ferroptosis inducers. Compounds have been classified based on their chemical structure, and modulation of ferroptosis-related biochemical pathways has been reported. Future Directions: The outcomes herein collected represent a fascinating starting point from which to take hints for future drug discovery studies aimed at identifying ferroptosis-inducing natural compounds for anticancer therapies. Antioxid. Redox Signal. 40, 40-85.
Collapse
Affiliation(s)
- Valeria Consoli
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | | | - Valeria Sorrenti
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Valeria Pittalà
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| | - Luca Vanella
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
- Department of Drug and Health Sciences, CERNUT—Research Centre on Nutraceuticals and Health Products, University of Catania, Catania, Italy
| |
Collapse
|
130
|
Hays SB, Corvino G, Lorié BD, McMichael WV, Mehdi SA, Rieser C, Rojas AE, Hogg ME. Prince and princesses: The current status of robotic surgery in surgical oncology. J Surg Oncol 2024; 129:164-182. [PMID: 38031870 DOI: 10.1002/jso.27536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 12/01/2023]
Abstract
Robotic surgery has experienced a dramatic increase in utilization across general surgery over the last two decades, including in surgical oncology. Although urologists and gynecologists were the first to show that this technology could be utilized in cancer surgery, the robot is now a powerful tool in the treatment of gastrointestinal, hepato-pancreatico-biliary, colorectal, endocrine, and soft tissue malignancies. While long-term outcomes are still pending, short-term outcomes have showed promise for this technologic advancement of cancer surgery.
Collapse
Affiliation(s)
- Sarah B Hays
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Gaetano Corvino
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Benjamin D Lorié
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - William V McMichael
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Syed A Mehdi
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Caroline Rieser
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Aram E Rojas
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Melissa E Hogg
- Department of Surgery, Evanston Hospital, NorthShore University HealthSystem, Evanston, Illinois, USA
| |
Collapse
|
131
|
Wei X, Ni J, Yuan L, Li X. Hematoporphyrin derivative photodynamic therapy induces apoptosis and suppresses the migration of human esophageal squamous cell carcinoma cells by regulating the PI3K/AKT/mTOR signaling pathway. Oncol Lett 2024; 27:17. [PMID: 38034489 PMCID: PMC10688503 DOI: 10.3892/ol.2023.14150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/02/2023] [Indexed: 12/02/2023] Open
Abstract
Esophageal cancer is one of the most common cancer types in humans worldwide. Photodynamic therapy (PDT) is a promising therapeutic strategy for the treatment of cancer. However, its underlying mechanism needs to be studied thoroughly. The present study focused on the antitumor effect and underlying mechanism of the use of hematoporphyrin derivative (HpD)-PDT against human esophageal squamous cell carcinoma cells via regulation of the PI3K/AKT/mTOR signaling pathway. A Cell Counting Kit-8 assay was used to measure cell viability. Migration was evaluated using a wound healing assay. An annexin V-FITC/PI kit was used to determine cell apoptosis rates. Protein expression levels were analyzed via western blotting. Reverse transcription-quantitative PCR was used to detect gene expression levels. A 2',7'-dichlorodihydrofluorescein diacetate kit was chosen to evaluate intracellular reactive oxygen species levels via flow cytometry. Cell viability and migration were decreased in KYSE-150 cells after HpD-PDT treatment. Cellular apoptosis was induced after HpD-PDT treatment, and the same trend was observed for autophagy. Furthermore, the PI3K/AKT/mTOR signaling pathway was inhibited. The viability and migration of KYSE-150 cells were significantly inhibited, and apoptosis was induced more effectively following treatment with a combination of HpD-PDT and the PI3K inhibitor, a final concentration of 20 µM LY294002. In conclusion, HpD-PDT could suppress esophageal cancer cell viability, induce apoptosis and inhibit migration by downregulating the PI3K/AKT/mTOR signaling pathway. Combination of HpD-PDT with PI3K inhibitor (LY294002) could enhance the therapeutic efficacy compared with that demonstrated by HpD-PDT alone. Further studies on combination therapy are required to achieve improved clinical outcomes.
Collapse
Affiliation(s)
- Xin Wei
- Department of Internal Medicine, First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Jinliang Ni
- Department of Internal Medicine, First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Lin Yuan
- Department of Internal Medicine, First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xueliang Li
- Department of Internal Medicine, First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
132
|
Nauser S, Steinkohl E, Olesen SS, Drewes AM, Frøkjær JB. Co-existence of hepatic and pancreatic fibrosis in chronic pancreatitis patients including associated risk factors: a magnetic resonance elastography study. Scand J Gastroenterol 2024; 59:100-107. [PMID: 37615331 DOI: 10.1080/00365521.2023.2250496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/15/2023] [Indexed: 08/25/2023]
Abstract
OBJECTIVES To investigate the co-existence of hepatic and pancreatic fibrosis using magnetic resonance elastography (MRE) in chronic pancreatitis (CP), including the association between hepatic and pancreatic MRE-derived stiffness and exploration of potential etiological risk factors. MATERIALS AND METHODS Fifty-four CP patients and 35 healthy controls underwent hepatic and pancreatic MRE with measurements of tissue stiffness. Clinical parameters including stage (probable or definite CP), etiology of CP, the presence of diabetes or exocrine insufficiency, and previous history of common bile duct stenosis were assessed. Uni- and multivariate regression models were used to investigate risk factors associated with hepatic fibrosis/stiffness in CP patients. RESULTS Fifteen percent of CP patients and none of the controls had abnormal liver stiffness (>2.5 kPa), p = 0.02. 5.6% of CP patients had liver stiffness indicating F1 fibrosis (>2.93 kPa). However, hepatic stiffness was not higher in patients than in healthy controls (2.20 ± 0.41 vs 2.08 ± 0.21 kPa, p = 0.10). In patients, a positive association was seen between hepatic and pancreatic stiffness (r = 0.270, p = 0.048). In the multivariate analysis (adjusted for age, gender and BMI), liver stiffness was significantly associated with alcoholic etiology of CP (p = 0.029). In contrast, stage of CP, history of common bile duct stenosis, and the presence of diabetes or exocrine insufficiency were not associated with liver stiffness (all p > 0.14). CONCLUSIONS Only a modest co-existence of hepatic and pancreatic fibrosis was observed in CP. However, the positive association between hepatic and pancreatic stiffness indicates some level of common pathophysiology. Especially, alcoholic etiology of CP was related to increased hepatic stiffness.
Collapse
Affiliation(s)
- Serena Nauser
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Emily Steinkohl
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
- Centre for Pancreatic Diseases, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Søren Schou Olesen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Centre for Pancreatic Diseases, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Asbjørn Mohr Drewes
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Centre for Pancreatic Diseases, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Jens Brøndum Frøkjær
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Radiology, Aalborg University Hospital, Aalborg, Denmark
| |
Collapse
|
133
|
Pandey P, Khan F, Singh M, Verma A, Kumar H, Mazumder A, Rakhra G. Study Deciphering the Crucial Involvement of Notch Signaling Pathway in Human Cancers. Endocr Metab Immune Disord Drug Targets 2024; 24:1241-1253. [PMID: 37997805 DOI: 10.2174/0118715303261691231107113548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 11/25/2023]
Abstract
In recent years, dysregulation of the notch pathway has been associated with the development and progression of various cancers. Notch signaling is involved in several cellular processes, such as proliferation, differentiation, apoptosis, and angiogenesis, and its abnormal activation can lead to uncontrolled cell growth and tumorigenesis. In various human cancers, the Notch pathway has been shown to have both tumor-promoting and tumor-suppressive effects, depending on the context and stage of cancer development. Notch signaling has been implicated in tumor initiation, cancer cell proliferation, cell migration and maintenance of cancer stem cells in several human cancers, including leukemia, breast, pancreatic and lung cancer. Understanding the role of the Notch pathway in cancer development and progression may provide new opportunities for the development of potent targeted therapies for cancer treatment. Several drugs targeting the Notch pathway are currently in preclinical or clinical development and may hold promise for anticancer therapy in the future.
Collapse
Affiliation(s)
- Pratibha Pandey
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Fahad Khan
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Megha Singh
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Aditi Verma
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Hariom Kumar
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, UP, India
| | - Avijit Mazumder
- Department of Pharmacology, Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, 201306, India
| | - Gurmeen Rakhra
- Department of Biochemistry, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
134
|
Yao B, Xing M, Meng S, Li S, Zhou J, Zhang M, Yang C, Qu S, Jin Y, Yuan H, Zen K, Ma C. EBF2 Links KMT2D-Mediated H3K4me1 to Suppress Pancreatic Cancer Progression via Upregulating KLLN. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302037. [PMID: 38015024 PMCID: PMC10787067 DOI: 10.1002/advs.202302037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/09/2023] [Indexed: 11/29/2023]
Abstract
Mono-methylation of histone H3 on Lys 4 (H3K4me1), which is catalyzed by histone-lysine N-methyltransferase 2D (KMT2D), serves as an important epigenetic regulator in transcriptional control. In this study, the authors identify early B-cell factor 2 (EBF2) as a binding protein of H3K4me1. Combining analyses of RNA-seq and ChIP-seq data, the authors further identify killin (KLLN) as a transcriptional target of KMT2D and EBF2 in pancreatic ductal adenocarcinoma (PDAC) cells. KMT2D-dependent H3K4me1 and EBF2 are predominantly over-lapped proximal to the transcription start site (TSS) of KLLN gene. Comprehensive functional assays show that KMT2D and EBF2 cooperatively inhibit PDAC cells proliferation, migration, and invasion through upregulating KLLN. Such inhibition on PDAC progression is also achieved through increasing H3K4me1 level by GSK-LSD1, a selective inhibitor of lysine-specific demethylase 1 (LSD1). Taken together, these findings reveal a new mechanism underlying PDAC progression and provide potential therapeutic targets for PDAC treatment.
Collapse
Affiliation(s)
- Bing Yao
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Mengying Xing
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Shixin Meng
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Shang Li
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Jingwan Zhou
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Ming Zhang
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Chen Yang
- The State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Shuang Qu
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Yucui Jin
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
| | - Hongyan Yuan
- Department of Oncology and Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDC20007USA
| | - Ke Zen
- The State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing University163 Xianlin AvenueNanjing210023China
| | - Changyan Ma
- Department of Medical GeneticsNanjing Medical University101 Longmian AvenueNanjing211166China
- Jiangsu Key Laboratory of XenotransplantationNanjing Medical University101 Longmian AvenueNanjing211166China
| |
Collapse
|
135
|
Gazzillo A, Volponi C, Soldani C, Polidoro MA, Franceschini B, Lleo A, Bonavita E, Donadon M. Cellular Senescence in Liver Cancer: How Dying Cells Become "Zombie" Enemies. Biomedicines 2023; 12:26. [PMID: 38275386 PMCID: PMC10813254 DOI: 10.3390/biomedicines12010026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Liver cancer represents the fourth leading cause of cancer-associated death worldwide. The heterogeneity of its tumor microenvironment (TME) is a major contributing factor of metastasis, relapse, and drug resistance. Regrettably, late diagnosis makes most liver cancer patients ineligible for surgery, and the frequent failure of non-surgical therapeutic options orientates clinical research to the investigation of new drugs. In this context, cellular senescence has been recently shown to play a pivotal role in the progression of chronic inflammatory liver diseases, ultimately leading to cancer. Moreover, the stem-like state triggered by senescence has been associated with the emergence of drug-resistant, aggressive tumor clones. In recent years, an increasing number of studies have emerged to investigate senescence-associated hepatocarcinogenesis and its derived therapies, leading to promising results. In this review, we intend to provide an overview of the recent evidence that unveils the role of cellular senescence in the most frequent forms of primary and metastatic liver cancer, focusing on the involvement of this mechanism in therapy resistance.
Collapse
Affiliation(s)
- Aurora Gazzillo
- Cellular and Molecular Oncoimmunology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.G.); (C.V.); (E.B.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy;
| | - Camilla Volponi
- Cellular and Molecular Oncoimmunology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.G.); (C.V.); (E.B.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy;
| | - Cristiana Soldani
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.S.); (M.A.P.); (B.F.)
| | - Michela Anna Polidoro
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.S.); (M.A.P.); (B.F.)
| | - Barbara Franceschini
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.S.); (M.A.P.); (B.F.)
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy;
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.S.); (M.A.P.); (B.F.)
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Eduardo Bonavita
- Cellular and Molecular Oncoimmunology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (A.G.); (C.V.); (E.B.)
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy;
| | - Matteo Donadon
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.S.); (M.A.P.); (B.F.)
- Department of Health Sciences, Università del Piemonte Orientale, 28100 Novara, Italy
- Department of General Surgery, University Maggiore Hospital della Carità, 28100 Novara, Italy
| |
Collapse
|
136
|
Jenkins SV, Jung S, Jamshidi-Parsian A, Borrelli MJ, Dings RPM, Griffin RJ. Morphological Effects and In Vitro Biological Mechanisms of Radiation-Induced Cell Killing by Gold Nanomaterials. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58241-58250. [PMID: 38059477 DOI: 10.1021/acsami.3c15358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Gold nanomaterials have been shown to augment radiation therapy both in vitro and in vivo. However, studies on these materials are mostly phenomenological due to nanoparticle heterogeneity and the complexity of biological systems. Even accurate quantification of the particle dose still results in bulk average biases; the effect on individual cells is not measured but rather the effect on the overall population. To perform quantitative nanobiology, we coated glass coverslips uniformly at varying densities with Au nanoparticle preparations with different morphologies (45 nm cages, 25 nm spheres, and 30 nm rods). Consequently, the effect of a specific number of particles per unit area in contact with breast cancer cells growing on the coated surfaces was ascertained. Gold nanocages showed the highest degree of radiosensitization on a per particle basis, followed by gold nanospheres and gold nanorods, respectively. All three materials showed little cytotoxic effect at 0 Gy, but clonogenic survival decreased proportionally with the radiation dose and particle coverage density. A similar trend was seen in vivo in the combined treatment antitumor response in 4T1 tumor-bearing animals. The presence of gold affected the type and quantity of reactive oxygen species generated, specifically superoxide and hydroxyl radicals, and the concentration of nanocages correlated with the development of more numerous double-stranded DNA breaks and increased protein oxidation as measured by carbonylation. This work demonstrates the dependence on morphology and concentration of radiation enhancement by gold nanomaterials and may lead to a novel method to differentiate intra- and extracellular functionalities of gold nanomedicine treatment strategies. It further provides insights that can guide the rational development of gold nanomaterial-based radiosensitizers for clinical use.
Collapse
Affiliation(s)
- Samir V Jenkins
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Seunghyun Jung
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Azemat Jamshidi-Parsian
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Michael J Borrelli
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Ruud P M Dings
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| | - Robert J Griffin
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas 72205, United States
| |
Collapse
|
137
|
Wang Q, Shen K, Fei B, Luo H, Li R, Wang Z, Wei M, Xie Z. A predictive model for early death in elderly colorectal cancer patients: a population-based study. Front Oncol 2023; 13:1278137. [PMID: 38173840 PMCID: PMC10764026 DOI: 10.3389/fonc.2023.1278137] [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: 08/15/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Purpose The purpose of this study is to determine what variables contribute to the early death of elderly colorectal cancer patients (ECRC) and to generate predictive nomograms for this population. Methods This retrospective cohort analysis included elderly individuals (≥75 years old) diagnosed with colorectal cancer (CRC) from 2010-2015 in the Surveillance, Epidemiology, and End Result databases (SEER) databases. The external validation was conducted using a sample of the Chinese population obtained from the China-Japan Union Hospital of Jilin University. Logistic regression analyses were used to ascertain variables associated with early death and to develop nomograms. The nomograms were internally and externally validated with the help of the receiver operating characteristic curve (ROC), calibration curve, and decision curve analysis (DCA). Results The SEER cohort consisted of 28,111 individuals, while the Chinese cohort contained 315 cases. Logistic regression analyses shown that race, marital status, tumor size, Grade, T stage, N stage, M stage, brain metastasis, liver metastasis, bone metastasis, surgery, chemotherapy, and radiotherapy were independent prognostic factors for all-cause and cancer-specific early death in ECRC patients; The variable of sex was only related to an increased risk of all-cause early death, whereas the factor of insurance status was solely associated with an increased risk of cancer-specific early death. Subsequently, two nomograms were devised to estimate the likelihood of all-cause and cancer-specific early death among individuals with ECRC. The nomograms exhibited robust predictive accuracy for predicting early death of ECRC patients, as evidenced by both internal and external validation. Conclusion We developed two easy-to-use nomograms to predicting the likelihood of early death in ECRC patients, which would contribute significantly to the improvement of clinical decision-making and the formulation of personalized treatment approaches for this particular population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Zhongshi Xie
- Department of Gastrointestinal Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
138
|
Niu J, Jiang W, Fan D, Li X, Zhou W, Zhang H. Research trends on immunotherapy for pancreatic cancer: A bibliometric analysis. Hum Vaccin Immunother 2023; 19:2269794. [PMID: 37885280 PMCID: PMC10760365 DOI: 10.1080/21645515.2023.2269794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
This study aims to summarize and visually analyze the current research status in pancreatic cancer immunotherapy during the past two decades by bibliometrics and explore the current research hotspots and future development directions. The literature related to pancreatic cancer immunotherapy from 2002 to 2021 was downloaded from the core database of the Web of Science. VOSviewer and CiteSpace software were used to visualize the included literature. A total of 2528 articles were included. In the past two decades, publications in the pancreatic cancer immunotherapy field have increased almost annually. As the country with the largest publications, the United States has various research institutions dedicated to pancreatic cancer immunotherapy. Jaffee EM and Zheng L from Johns Hopkins University and Vonderheide RH from the University of Pennsylvania have published the most articles in this field. The current research hotspots of pancreatic cancer immunotherapy include the tumor microenvironment, immune cells, immune checkpoint blockade, and combination therapy. The study of novel immunotherapies and combination therapy may become the primary focus of future research on pancreatic cancer immunotherapy. More prospective clinical studies with high evidence levels should be conducted.
Collapse
Affiliation(s)
- Jubao Niu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wenkai Jiang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongao Fan
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Xin Li
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wence Zhou
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Hui Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
139
|
Zirakchian Zadeh M. PET/CT in assessment of colorectal liver metastases: a comprehensive review with emphasis on 18F-FDG. Clin Exp Metastasis 2023; 40:465-491. [PMID: 37682423 DOI: 10.1007/s10585-023-10231-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/21/2023] [Indexed: 09/09/2023]
Abstract
Approximately 25% of those who are diagnosed with colorectal cancer will develop colorectal liver metastases (CRLM) as their illness advances. Despite major improvements in both diagnostic and treatment methods, the prognosis for patients with CRLM is still poor, with low survival rates. Accurate employment of imaging methods is critical in identifying the most effective treatment approach for CRLM. Different imaging modalities are used to evaluate CRLM, including positron emission tomography (PET)/computed tomography (CT). Among the PET radiotracers, fluoro-18-deoxyglucose (18F-FDG), a glucose analog, is commonly used as the primary radiotracer in assessment of CRLM. As the importance of 18F-FDG-PET/CT continues to grow in assessment of CRLM, developing a comprehensive understanding of this subject becomes imperative for healthcare professionals from diverse disciplines. The primary aim of this article is to offer a simplified and comprehensive explanation of PET/CT in the evaluation of CRLM, with a deliberate effort to minimize the use of technical nuclear medicine terminology. This approach intends to provide various healthcare professionals and researchers with a thorough understanding of the subject matter.
Collapse
Affiliation(s)
- Mahdi Zirakchian Zadeh
- Molecular Imaging and Therapy and Interventional Radiology Services, Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
140
|
Zhang Z, Zhao Y, Wang Y, Zhao Y, Guo J. Autophagy/ferroptosis in colorectal cancer: Carcinogenic view and nanoparticle-mediated cell death regulation. ENVIRONMENTAL RESEARCH 2023; 238:117006. [PMID: 37669735 DOI: 10.1016/j.envres.2023.117006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/19/2023] [Accepted: 08/26/2023] [Indexed: 09/07/2023]
Abstract
The cell death mechanisms have a long history of being evaluated in diseases and pathological events. The ability of triggering cell death is considered to be a promising strategy in cancer therapy, but some mechanisms have dual functions in cancer, requiring more elucidation of underlying factors. Colorectal cancer (CRC) is a disease and malignant condition of colon and rectal that causes high mortality and morbidity. The autophagy targeting in CRC is therapeutic importance and this cell death mechanism can interact with apoptosis in inhibiting or increasing apoptosis. Autophagy has interaction with ferroptosis as another cell death pathway in CRC and can accelerate ferroptosis in suppressing growth and invasion. The dysregulation of autophagy affects the drug resistance in CRC and pro-survival autophagy can induce drug resistance. Therefore, inhibition of protective autophagy enhances chemosensitivity in CRC cells. Moreover, autophagy displays interaction with metastasis and EMT as a potent regulator of invasion in CRC cells. The same is true for ferroptosis, but the difference is that function of ferroptosis is determined and it can reduce viability. The lack of ferroptosis can cause development of chemoresistance in CRC cells and this cell death mechanism is regulated by various pathways and mechanisms that autophagy is among them. Therefore, current review paper provides a state-of-art analysis of autophagy, ferroptosis and their crosstalk in CRC. The nanoparticle-mediated regulation of cell death mechanisms in CRC causes changes in progression. The stimulation of ferroptosis and control of autophagy (induction or inhibition) by nanoparticles can impair CRC progression. The engineering part of nanoparticle synthesis to control autophagy and ferroptosis in CRC still requires more attention.
Collapse
Affiliation(s)
- Zhibin Zhang
- Chengde Medical College, College of Traditional Chinese Medicine, Chengde, Hebei, 067000, China.
| | - Yintao Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yuman Wang
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Yutang Zhao
- Chengde Medical College, Chengde, Hebei, 067000, China
| | - Jianen Guo
- Chengde Medical College, Chengde, Hebei, 067000, China
| |
Collapse
|
141
|
Gerwing M, Schindler P, Katou S, Köhler M, Stamm AC, Schmidt VF, Heindel W, Struecker B, Morgul H, Pascher A, Wildgruber M, Masthoff M. Multi-organ Radiomics-Based Prediction of Future Remnant Liver Hypertrophy Following Portal Vein Embolization. Ann Surg Oncol 2023; 30:7976-7985. [PMID: 37670120 PMCID: PMC10625940 DOI: 10.1245/s10434-023-14241-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 07/24/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Portal vein embolization (PVE) is used to induce remnant liver hypertrophy prior to major hepatectomy. The purpose of this study was to evaluate the predictive value of baseline computed tomography (CT) data for future remnant liver (FRL) hypertrophy after PVE. METHODS In this retrospective study, all consecutive patients undergoing right-sided PVE with or without hepatic vein embolization between 2018 and 2021 were included. CT volumetry was performed before and after PVE to assess standardized FRL volume (sFRLV). Radiomic features were extracted from baseline CT after segmenting liver (without tumor), spleen and bone marrow. For selecting features that allow classification of response (hypertrophy ≥ 1.33), a stepwise dimension reduction was performed. Logistic regression models were fitted and selected features were tested for their predictive value. Decision curve analysis was performed on the test dataset. RESULTS A total of 53 patients with liver tumor were included in this study. sFRLV increased significantly after PVE, with a mean hypertrophy of FRL of 1.5 ± 0.3-fold. sFRLV hypertrophy ≥ 1.33 was reached in 35 (66%) patients. Three independent radiomic features, i.e. liver-, spleen- and bone marrow-associated, differentiated well between responders and non-responders. A logistic regression model revealed the highest accuracy (area under the curve 0.875) for the prediction of response, with sensitivity of 1.0 and specificity of 0.5. Decision curve analysis revealed a positive net benefit when applying the model. CONCLUSIONS This proof-of-concept study provides first evidence of a potential predictive value of baseline multi-organ radiomics CT data for FRL hypertrophy after PVE.
Collapse
Affiliation(s)
- Mirjam Gerwing
- Clinic for Radiology, University Hospital Münster, Münster, Germany.
| | | | - Shadi Katou
- Department for General, Visceral and Transplant Surgery, University Hospital Münster, Münster, Germany
| | - Michael Köhler
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | | | | | - Walter Heindel
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| | - Benjamin Struecker
- Department for General, Visceral and Transplant Surgery, University Hospital Münster, Münster, Germany
| | - Haluk Morgul
- Department for General, Visceral and Transplant Surgery, University Hospital Münster, Münster, Germany
| | - Andreas Pascher
- Department for General, Visceral and Transplant Surgery, University Hospital Münster, Münster, Germany
| | - Moritz Wildgruber
- Clinic for Radiology, University Hospital Münster, Münster, Germany
- Department for Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Max Masthoff
- Clinic for Radiology, University Hospital Münster, Münster, Germany
| |
Collapse
|
142
|
Xu L, Ma S, Fan B, Yuan Z, Yin P. Bufalin-loaded vitamin E succinate-grafted chitosan oligosaccharide/RGD-conjugated TPGS mixed micelles inhibit intraperitoneal metastasis of ovarian cancer. Cancer Nanotechnol 2023. [DOI: 10.1186/s12645-023-00178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Abstract
Background
Intraperitoneal metastasis is one of the major causes of the high mortality rate of ovarian cancer. Bufalin (BU) is an effective component of the traditional Chinese medicine Chansu that exerts antitumor effects, including metastasis inhibition. In our previous studies, we found that BU inhibited the migration and invasion of ovarian cancer cells. However, the application of BU is limited due to its insolubility, toxicity and imprecise targeting. The aim of this study was to use vitamin E succinate (VES)-grafted chitosan oligosaccharide (CSO)/arginine-glycine-aspartic acid peptide (RGD)-conjugated d-alpha-tocopheryl polyethylene glycol 1000 succinate (TPGS) mixed micelles (VeC/T-RGD MMs) to deliver BU to ovarian cancer cells to inhibit intraperitoneal metastasis. Moreover, the toxicity of BU was reduced by coating it with the mixed micelles to increase its biocompatibility for practical applications.
Results
The BU-loaded VeC/T-RGD MMs (BU@MMs) had an average diameter of 161 ± 1.4 nm, a zeta potential of 4.49 ± 1.54 mV and a loading efficiency of 2.54%. The results showed that these micelles inhibited cell proliferation, induced apoptosis, and reduced the migration and invasion of A2780 and SKOV3 cells. Further studies indicated that BU@MMs enhanced the levels of e-cadherin and decreased the expression levels of N-cadherin, vimentin and Snail in vitro. In addition, the mixed micelles effectively enhanced the anticancer effect and inhibited intraperitoneal metastasis in intraperitoneal metastatic models. The BU@MMs exhibited fewer toxic side effects than BU, indicating better biocompatibility and biosafety for in vivo applications.
Conclusions
Our studies show that BU@MMs are a potential multifunctional nano-drug delivery system that can effectively inhibit the intraperitoneal metastasis of ovarian cancer.
Collapse
|
143
|
Zwart MJ, van den Broek B, de Graaf N, Suurmeijer JA, Augustinus S, te Riele WW, van Santvoort HC, Hagendoorn J, Borel Rinkes IH, van Dam JL, Takagi K, Tran KT, Schreinemakers J, van der Schelling G, Wijsman JH, de Wilde RF, Festen S, Daams F, Luyer MD, de Hingh IH, Mieog JS, Bonsing BA, Lips DJ, Abu Hilal M, Busch OR, Saint-Marc O, Zeh HJ, Zureikat AH, Hogg ME, Koerkamp BG, Molenaar IQ, Besselink MG. The Feasibility, Proficiency, and Mastery Learning Curves in 635 Robotic Pancreatoduodenectomies Following a Multicenter Training Program: "Standing on the Shoulders of Giants". Ann Surg 2023; 278:e1232-e1241. [PMID: 37288547 PMCID: PMC10631507 DOI: 10.1097/sla.0000000000005928] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
OBJECTIVE To assess the feasibility, proficiency, and mastery learning curves for robotic pancreatoduodenectomy (RPD) in "second-generation" RPD centers following a multicenter training program adhering to the IDEAL framework. BACKGROUND The long learning curves for RPD reported from "pioneering" expert centers may discourage centers interested in starting an RPD program. However, the feasibility, proficiency, and mastery learning curves may be shorter in "second-generation" centers that participated in dedicated RPD training programs, although data are lacking. We report on the learning curves for RPD in "second-generation" centers trained in a dedicated nationwide program. METHODS Post hoc analysis of all consecutive patients undergoing RPD in 7 centers that participated in the LAELAPS-3 training program, each with a minimum annual volume of 50 pancreatoduodenectomies, using the mandatory Dutch Pancreatic Cancer Audit (March 2016-December 2021). Cumulative sum analysis determined cutoffs for the 3 learning curves: operative time for the feasibility (1) risk-adjusted major complication (Clavien-Dindo grade ≥III) for the proficiency, (2) and textbook outcome for the mastery, (3) learning curve. Outcomes before and after the cutoffs were compared for the proficiency and mastery learning curves. A survey was used to assess changes in practice and the most valued "lessons learned." RESULTS Overall, 635 RPD were performed by 17 trained surgeons, with a conversion rate of 6.6% (n=42). The median annual volume of RPD per center was 22.5±6.8. From 2016 to 2021, the nationwide annual use of RPD increased from 0% to 23% whereas the use of laparoscopic pancreatoduodenectomy decreased from 15% to 0%. The rate of major complications was 36.9% (n=234), surgical site infection 6.3% (n=40), postoperative pancreatic fistula (grade B/C) 26.9% (n=171), and 30-day/in-hospital mortality 3.5% (n=22). Cutoffs for the feasibility, proficiency, and mastery learning curves were reached at 15, 62, and 84 RPD. Major morbidity and 30-day/in-hospital mortality did not differ significantly before and after the cutoffs for the proficiency and mastery learning curves. Previous experience in laparoscopic pancreatoduodenectomy shortened the feasibility (-12 RPDs, -44%), proficiency (-32 RPDs, -34%), and mastery phase learning curve (-34 RPDs, -23%), but did not improve clinical outcome. CONCLUSIONS The feasibility, proficiency, and mastery learning curves for RPD at 15, 62, and 84 procedures in "second-generation" centers after a multicenter training program were considerably shorter than previously reported from "pioneering" expert centers. The learning curve cutoffs and prior laparoscopic experience did not impact major morbidity and mortality. These findings demonstrate the safety and value of a nationwide training program for RPD in centers with sufficient volume.
Collapse
Affiliation(s)
- Maurice J.W. Zwart
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Bram van den Broek
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Nine de Graaf
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
- Department of Surgery, Fondazione Poliambulanza Institute, Brescia, Italy
| | - José A. Suurmeijer
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Simone Augustinus
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Wouter W. te Riele
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St Antonius Hospital Nieuwegein, Utrecht University, Utrecht, the Netherlands
| | - Hjalmar C. van Santvoort
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St Antonius Hospital Nieuwegein, Utrecht University, Utrecht, the Netherlands
| | - Jeroen Hagendoorn
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St Antonius Hospital Nieuwegein, Utrecht University, Utrecht, the Netherlands
| | - Inne H.M. Borel Rinkes
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St Antonius Hospital Nieuwegein, Utrecht University, Utrecht, the Netherlands
| | - Jacob L. van Dam
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Kosei Takagi
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Khé T.C. Tran
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | | | - Jan H. Wijsman
- Department of Surgery, Amphia Medical Center, Breda, the Netherlands
| | - Roeland F. de Wilde
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Freek Daams
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Misha D. Luyer
- Department of Surgery, Catharina Hospital, Eindhoven, the Netherlands
| | | | - Jan S.D. Mieog
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Bert A. Bonsing
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Daan J. Lips
- Department of Surgery, Twente Medical Spectrum, Enschede, the Netherlands
| | - Mohamed Abu Hilal
- Department of Surgery, Fondazione Poliambulanza Institute, Brescia, Italy
- Department of Surgery, Southampton University Hospital NHS Foundation Trust, Southampton, UK
| | - Olivier R. Busch
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| | | | - Herbert J. Zeh
- Department of Surgery, University of Texas, Southwestern, Dallas, TX
| | - Amer H. Zureikat
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Melissa E. Hogg
- Department of Surgery, Northshore University HealthSystem, Chicago, IL
| | - Bas G. Koerkamp
- Department of Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Isaac Q. Molenaar
- Department of Surgery, Regional Academic Cancer Center Utrecht, UMC Utrecht Cancer Center & St Antonius Hospital Nieuwegein, Utrecht University, Utrecht, the Netherlands
| | - Marc G. Besselink
- Department of Surgery, Amsterdam UMC, location University of Amsterdam, Amsterdam, the Netherlands
- Cancer Center Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
144
|
Yue CF, Chen JG, Li ZY, Li LS, Chen JR, Xie HX, Zhang B, Guo YM. Tumor cell-derived exosomes mediating hsa_circ_0001739/lncRNA AC159540.1 facilitate liver metastasis in colorectal cancer. Cell Biol Toxicol 2023; 39:2551-2568. [PMID: 37957486 DOI: 10.1007/s10565-023-09837-2] [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: 03/06/2022] [Accepted: 10/25/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND The current study probed into how tumor cell-derived exosomes (Exos) mediated hsa_circ_0001739/lncRNA AC159540.1 to manipulate microRNA (miR)-218-5p/FTO-N6-methyladenosine (m6A)/MYC signal axis in liver metastasis in colorectal cancer (CRC). METHODS hsa_circ_0001739 and lncRNA AC159540.1 were identified as the upstream regulator of miR-218-5p using ENCORI and LncBase databases. Expression patterns of miR-218-5p, hsa_circ_0001739, lncRNA AC159540.1, FTO, and MYC were detected, accompanied by loss-and-gain-of function assays to examine their effects on CRC cell biological functions. SW480 cells-derived Exos were purified, followed by in vitro studies to uncover the effect of hsa_circ_0001739/lncRNA AC159540. RESULTS miR-218-5p was downregulated while hsa_circ_0001739/lncRNA AC159540.1 was upregulated in CRC tissues and cells. Silencing of hsa_circ_0001739/lncRNA AC159540.1 restrained the malignant phenotypes of CRC cells. Exos-mediated hsa_circ_0001739/lncRNA AC159540.1 competitively inhibited miR-218-5p to elevate FTO and MYC. The inducing role of Exos-mediated hsa_circ_0001739/lncRNA AC159540.1 in CRC was also validated in vivo. CONCLUSION Conclusively, Exos-mediated circ_0001739/lncRNA AC159540.1 regulatory network is critical for CRC, offering a theoretical basis for CRC treatment.
Collapse
Affiliation(s)
- Cai-Feng Yue
- Department of Laboratory Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, 524045, People's Republic of China
| | - Ju-Gao Chen
- Department of Oncology, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People's Republic of China
| | - Zi-Yue Li
- Prenatal Diagnostic Center and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Lai-Sheng Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Jie-Rong Chen
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, People's Republic of China
| | - Hai-Xia Xie
- Department of Laboratory Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, 524045, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Diagnosis, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, 524045, People's Republic of China
| | - Yun-Miao Guo
- Zhanjiang Institute of Clinical Medicine, Central People's Hospital of Zhanjiang, Guangdong Medical University Zhanjiang Central Hospital, Zhanjiang, 524045, People's Republic of China.
| |
Collapse
|
145
|
Song J, Liu Q, Han L, Song T, Huang S, Zhang X, He Q, Liang C, Zhu S, Xiong B. Hsa_circ_0009092/miR-665/NLK signaling axis suppresses colorectal cancer progression via recruiting TAMs in the tumor microenvironment. J Exp Clin Cancer Res 2023; 42:319. [PMID: 38008713 PMCID: PMC10680284 DOI: 10.1186/s13046-023-02887-8] [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: 08/01/2023] [Accepted: 11/06/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND It has been demonstrated that circularRNA (circRNAs) plays a critical role in various cancers. While the potential molecular mechanism of circRNAs in the progression of colorectal cancer (CRC) remains uncertain. METHODS Differentially expressed circRNAs were identified by RNA sequencing. RT-qPCR detected the expression of circ_0009092, miR-665, and NLK in CRC tissues and cells. Functions of circ_0009092 on tumor cell proliferation, migration, and invasion were investigated by a series of in vitro assays. The underlying mechanism of circ_0009092 was explored by bioinformatics analysis, RNA immunoprecipitation (RIP) and luciferase assays. A co-culture assay in vitro was performed to detect the affection of circ_0009092 on macrophage recruitment in the tumor microenvironment (TME). A xenograft mouse model was used to explore the effect of circ_0009092 on tumor growth. RESULTS Circ_0009092 was downregulated in CRCand predicted a good prognosis. Overexpression of circ_0009092 reduced tumor cell EMT, proliferation, migration, and invasion in vitro and in vivo. Mechanistically, circ_0009092 elevated the NLK expression via sponging miR-665 and suppressed the Wnt/β-catenin signaling pathway. EIF4EA3 induced circ_0009092 expression in CRC cells. In addition, NLK regulates phosphorylation and O-GlcNAcylation of STAT3 by binding to STAT3, thereby inhibiting CCL2 expression, in which it inhibits macrophage recruitment in the tumor microenvironment (TME). CONCLUSION EIF4A3 suppressed circ_0009092 biogenesis, whichinhibits CRC progression by sponging miR-665 to downregulate NLK. Circ_0009092/miR-665/NLK suppressed tumor EMT, proliferation, migration, and invasion by acting on the Wnt/β-catenin signaling pathway. NLK directly interacted with STAT3 and decreased the CCL2 expression, inhibiting the recruitment of tumor-associated macrophages (TAMs) in the TME. Our study provided novel insights into the roles of circ_0009092 as a novel promising prognostic and therapeutic target in CRC.
Collapse
Affiliation(s)
- Jialin Song
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Qing Liu
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lei Han
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Tiantian Song
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Sihao Huang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Xinyao Zhang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Qiuming He
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Chenxi Liang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China
| | - Shuai Zhu
- Department of Pancreatic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Bin Xiong
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Key Laboratory of Tumour Biological Behaviours, Wuhan, 430071, China.
| |
Collapse
|
146
|
Ma B, Zhao Y, Liu X, Huo M, Wang J, Ma J, Zhang Y, Qin C. Key Modulation of ROS and HSP for Effective Therapy Against Hypoxic Tumor with Multifunctional Nanosystem. Int J Nanomedicine 2023; 18:6829-6846. [PMID: 38026539 PMCID: PMC10664717 DOI: 10.2147/ijn.s432928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Background Though nanomedicine-based photothermal therapy (PTT) has demonstrated promising prospect in tumor treatment due to its high therapeutic efficiency and controllable range, the overexpression of heat shock proteins (HSPs) during PTT can lead to intracellular thermal resistance and reduce its effectiveness. Reactive oxygen species (ROS), followed by the application of chemodynamic therapy (CDT) and photodynamic therapy (PDT), can eliminate HSPs and overcome thermal resistance. However, the tumor microenvironment, including hypoxia and glutathione (GSH) overexpression, impedes the production of ROS and therapeutic efficacy of CDT and PDT. Therefore, we proposed a multifunctional nanoplatform (HMPB@TCPP-Cu) driving PTT/ PDT/ CDT synergistic therapy for tumor treatment via modulating ROS and HSPs. Methods and Results In this work, a novel nanoplatform (HMPB@TCPP-Cu) composed of O2/PTT supplier HMPB (hollow mesoporous Prussian blue) and the loaded PDT/CDT agent (TCPP-Cu2+) was prepared. HMPB acts as an photothermal converter, effectively raising the tumor temperature and inducing apoptosis. HMPB is also a potent catalase-like nanozyme, which can catalyze hydrogen peroxide into oxygen and reduce tumor hypoxia, thus elevating the efficiency of ROS production and the effectiveness of PDT with the wing of sonosensitizer-TCPP. The intracellular glutathione(GSH) was depleted by Cu2+ and •OH was generated along with the Cu2+/Cu+ converting and Cu+-mediated Fenton-like reaction. Subsequently, the increased levels of ROS effectively eliminate intratumoral thermal resistance. The HMPB@TCPP-Cu has achieved synergistic PTT/PDT/CDT for hepatoblastoma treatment and significant inhibition of tumor growth was detected both in vitro and in vivo. Conclusion This study presents a multifunctional nanoplatform that combines photothermal/ chemodynamic/ photodynamic therapy for efficient hepatoblastoma treatment via modulating ROS and HSPs. Collectively, this study provides an appealing strategy in the cleavage of thermal resistance and a novel assistance and enhancement on thermal-related therapies.
Collapse
Affiliation(s)
- Bangzhen Ma
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Yisheng Zhao
- School of Pharmaceutical Sciences, Key Laboratory for Natural Active Pharmaceutical Constituents Research in Universities of Shandong Province, Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, 250353, People’s Republic of China
| | - Xiaoli Liu
- Department of Hernia and Abdominal Wall Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100043, People’s Republic of China
| | - Mengping Huo
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Jinghong Wang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Jiwei Ma
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Yang Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| | - Chengkun Qin
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, People’s Republic of China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, People’s Republic of China
| |
Collapse
|
147
|
Arguello-Sánchez R, López-Callejas R, Rodríguez-Méndez BG, Scougall-Vilchis R, Velázquez-Enríquez U, Mercado-Cabrera A, Peña-Eguiluz R, Valencia-Alvarado R, Medina-Solís CE. Innovative Curved-Tip Reactor for Non-Thermal Plasma and Plasma-Treated Water Generation: Synergistic Impact Comparison with Sodium Hypochlorite in Dental Root Canal Disinfection. MATERIALS (BASEL, SWITZERLAND) 2023; 16:7204. [PMID: 38005133 PMCID: PMC10672626 DOI: 10.3390/ma16227204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/26/2023]
Abstract
Non-thermal plasmas (NTPs), known as cold atmospheric plasmas (CAPs), hold great potential for diverse medical applications, including dentistry. However, traditional linear and rigid dielectric barrier discharge reactors used for NTP generation encounter limitations in accessing oral cavities and root canals. To address this issue, we have developed an innovative NTP reactor featuring an angled end for improved accessibility. The central copper electrode, with a 0.59 mm diameter and adjustable length for desired angulation, is coated with zircon powder (ZrSiO4) to ensure stable NTP generation. This central electrode is housed within a stainless steel tube (3 mm internal diameter, 8 mm external diameter, and 100 mm length) with a 27° angle at one end, making it ergonomically suitable for oral applications. NTP generation involves polarizing the reactor electrodes with 13.56 MHz radio frequency signals, using helium gas as a working medium. We introduce plasma-treated water (PTW) as an adjunctive therapy to enhance biofilm eradication within root canals. A synergistic approach combining NTP and PTW is employed and compared to the gold standard (sodium hypochlorite, NaOCl), effectively neutralizing Enterococcus faecalis bacteria, even in scenarios involving biofilms. Moreover, applying NTP in both gaseous and liquid environments successfully achieves bacterial inactivation at varying treatment durations, demonstrating the device's suitability for medical use in treating root canal biofilms. The proposed NTP reactor, characterized by its innovative design, offers a practical and specific approach to plasma treatment in dental applications. It holds promise in combatting bacterial infections in root canals and oral cavities.
Collapse
Affiliation(s)
- Raúl Arguello-Sánchez
- Dental Reseach Center and Advanced Studies "Dr. Keisaburo Miyata", School of Dentistry, Autonomous University of Mexico State, Av. Paseo Tollocan, 13 Universidad, Toluca de Lerdo 50130, Mexico
| | - Régulo López-Callejas
- Department of Physics, National Institute for Nuclear Research, Carretera Mexico-Toluca S/N, Ocoyoacac 52750, Mexico
| | | | - Rogelio Scougall-Vilchis
- Dental Reseach Center and Advanced Studies "Dr. Keisaburo Miyata", School of Dentistry, Autonomous University of Mexico State, Av. Paseo Tollocan, 13 Universidad, Toluca de Lerdo 50130, Mexico
| | - Ulises Velázquez-Enríquez
- Dental Reseach Center and Advanced Studies "Dr. Keisaburo Miyata", School of Dentistry, Autonomous University of Mexico State, Av. Paseo Tollocan, 13 Universidad, Toluca de Lerdo 50130, Mexico
| | - Antonio Mercado-Cabrera
- Department of Physics, National Institute for Nuclear Research, Carretera Mexico-Toluca S/N, Ocoyoacac 52750, Mexico
| | - Rosendo Peña-Eguiluz
- Department of Physics, National Institute for Nuclear Research, Carretera Mexico-Toluca S/N, Ocoyoacac 52750, Mexico
| | - Raúl Valencia-Alvarado
- Department of Physics, National Institute for Nuclear Research, Carretera Mexico-Toluca S/N, Ocoyoacac 52750, Mexico
| | - Carlo Eduardo Medina-Solís
- Dental Reseach Center and Advanced Studies "Dr. Keisaburo Miyata", School of Dentistry, Autonomous University of Mexico State, Av. Paseo Tollocan, 13 Universidad, Toluca de Lerdo 50130, Mexico
- Dentistry Academic Area of the Health Sciences Institute, Autonomous University of Hidalgo State, Exhacienda de la Concepción S/N Carretera Actopan-Tilcuautla, San Agustin Tlaxiaca 42160, Mexico
| |
Collapse
|
148
|
Chen B, Moin A, Virk HUH, Jneid H, Virani SS, Krittanawong C. Association of Cardiovascular Disease and Pancreatitis: What Came First, the Chicken or the Egg? J Clin Med 2023; 12:7101. [PMID: 38002718 PMCID: PMC10672425 DOI: 10.3390/jcm12227101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/04/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: Recent studies suggest an association between pancreatitis and cardiovascular disease. This article aims to review the available evidence linking cardiovascular disease with acute and chronic pancreatitis. (2) Methods: A comprehensive search was conducted on the PubMed/MEDLINE database from inception to April 2022 using Medical Subject Heading and keywords related to pancreatitis and cardiovascular disease. The search was limited to English-language literature involving human subjects, and various study types, including observational studies, case-control studies, cohort studies, and clinical trials, were screened for eligibility. Following data extraction, the authors conducted a narrative synthesis of the studies. (3) Results: Longitudinal studies indicate that a history of acute pancreatitis is associated with an increased risk of acute atherosclerotic cardiovascular disease and acute coronary syndrome. Elevated triglyceride levels (>2000 mg/dL) have a temporal relationship with acute pancreatitis. Cross-sectional studies have shown that acute pancreatitis is associated with cardiac injury during the acute phase. Based on longitudinal studies, chronic pancreatitis is associated with an increased risk of cerebrovascular diseases. However, data regarding the relationship between chronic pancreatitis and myocardial infarction are conflicting. (4) Conclusions: Based on the available evidence, having a history of acute pancreatitis appears to increase the risk of acute atherosclerotic cardiovascular disease. However, there is insufficient evidence to conclude whether chronic pancreatitis is associated with cardiovascular disease, and no definitive studies have yielded conflicting results.
Collapse
Affiliation(s)
- Bing Chen
- Department of Gastroenterology and Hepatology, Geisinger Medical Center, Danville, PA 17822, USA
| | - Aleena Moin
- Department of Internal Medicine-Pediatrics, Geisinger Medical Center, Danville, PA 17822, USA
| | - Hafeez Ul Hassan Virk
- Harrington Heart & Vascular Institute, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Hani Jneid
- John Sealy Distinguished Centennial Chair in Cardiology, Division of Cardiology, University of Texas Medical Branch, Houston, TX 77058, USA
| | - Salim S. Virani
- Section of Cardiology and Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Office of the Vice Provost (Research), The Aga Khan University, Karachi 74800, Pakistan
| | - Chayakrit Krittanawong
- Cardiology Division, NYU Langone Health and NYU School of Medicine, New York, NY 10016, USA
| |
Collapse
|
149
|
Boanova LG, Altmayer S, Watte G, Raupp AA, Francisco MZ, De Oliveira GS, Hochhegger B, Andrade RGF. Detection of Liver Lesions in Colorectal Cancer Patients Using 18F-FDG PET/CT Dual-Time-Point Scan Imaging. Cancers (Basel) 2023; 15:5403. [PMID: 38001662 PMCID: PMC10670707 DOI: 10.3390/cancers15225403] [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: 09/25/2023] [Revised: 10/24/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
OBJECTIVE The aim of this study was to evaluate the diagnostic performance of dual-time-point fluorine-18-fluorodeoxyglucose positron emission computed tomography/computed tomography (18F-FDG PET/CT) compared to conventional early imaging for detecting colorectal liver metastases (CRLM) in colorectal cancer (CRC) patients. METHODS One hundred twenty-four consecutive CRC patients underwent dual-time-point imaging scans on a retrospective basis. Histopathological confirmation and/or clinical follow-up were accepted as the gold standard. Standard uptake values (SUV), signal-to-noise ratio (SNR), retention index (RI), tumor-to-normal liver ratio (TNR), and lesion sizes were measured for early and delayed PET scans. The diagnostic performance of early and delayed images was calculated on a per-patient basis and compared using McNemar's test. RESULTS Among the 124 patients, 57 (46%) had CRLM, 6 (4.8%) had benign lesions, and 61 (49.2%) had no concerning lesions detected. Smaller CRLM lesions (<5 cm3) showed significantly higher uptake in the delayed scans relative to early imaging (p < 0.001). The SUV and TNR increased significantly in delayed imaging of all metastatic lesions (p < 0.001). The retention index of all CRLM was high (40.8%), especially for small lesions (54.8%). A total of 177 lesions in delayed images and 124 in standard early images were identified. In a per-patient analysis, delayed imaging had significantly higher sensitivity (100% vs. 87.7%) and specificity (91.0% vs. 94.0%) compared to early imaging (p-value = 0.04). CONCLUSIONS The detection of liver lesions using dual-time-point PET/CT scan improves the sensitivity and specificity for the detection of colorectal liver metastasis.
Collapse
Affiliation(s)
- Luciane G. Boanova
- Faculty of Medicine, Pontificial Catholic University of Rio Grande do Sul, Av. Ipiranga 6690, Porto Alegre 90619-900, Brazil (B.H.)
- Department of Nuclear Medicine, Hospital Mae de Deus, Av. Jose de Alencar 286, Porto Alegre 90880-481, Brazil;
| | - Stephan Altmayer
- Faculty of Medicine, Pontificial Catholic University of Rio Grande do Sul, Av. Ipiranga 6690, Porto Alegre 90619-900, Brazil (B.H.)
| | - Guilherme Watte
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre, Rua Sarmento Leite 245, Porto Alegre 90050-170, Brazil; (G.W.); (M.Z.F.)
| | - Ana Amelia Raupp
- Department of Nuclear Medicine, Hospital Mae de Deus, Av. Jose de Alencar 286, Porto Alegre 90880-481, Brazil;
| | - Martina Zaguini Francisco
- Graduate Program in Pathology, Federal University of Health Sciences of Porto Alegre, Rua Sarmento Leite 245, Porto Alegre 90050-170, Brazil; (G.W.); (M.Z.F.)
| | - Guilherme Strieder De Oliveira
- School of Medicine, Federal University of Rio Grande do Sul, R. Ramiro Barcelos, 2400—Santa Cecília, Porto Alegre 90035-003, Brazil;
| | - Bruno Hochhegger
- Faculty of Medicine, Pontificial Catholic University of Rio Grande do Sul, Av. Ipiranga 6690, Porto Alegre 90619-900, Brazil (B.H.)
| | - Rubens G. F. Andrade
- Faculty of Medicine, Pontificial Catholic University of Rio Grande do Sul, Av. Ipiranga 6690, Porto Alegre 90619-900, Brazil (B.H.)
| |
Collapse
|
150
|
Angeli-Pahim I, Chambers A, Duarte S, Zarrinpar A. Current Trends in Surgical Management of Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:5378. [PMID: 38001637 PMCID: PMC10670586 DOI: 10.3390/cancers15225378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 10/16/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide. Surgical management, including hepatic resection, liver transplantation, and ablation, offers the greatest potential for a curative approach. This review aims to discuss recent advancements in HCC surgery and identify unresolved issues in the field. Treatment selection relies on the BCLC staging system, with surgical therapies primarily recommended for early-stage disease. Recent studies have shown that patients previously considered unresectable, such as those with portal vein tumor thrombus and uncomplicated portal hypertension, may benefit from hepatic resection. Minimally invasive surgery and improved visualization techniques are also explored, alongside new techniques for optimizing future liver remnant, ex vivo resection, and advancements in hemorrhage control. Liver transplantation criteria, particularly the long-standing Milan criteria, are critically examined. Alternative criteria proposed and tested in specific regions are presented. In the context of organ shortage, bridging therapy plays a critical role in preventing tumor progression and maintaining patients eligible for transplantation. Lastly, we explore emerging ablation modalities, comparing them with the current standard, radiofrequency ablation. In conclusion, this comprehensive review provides insights into recent trends and future prospects in the surgical management of HCC, highlighting areas that require further investigation.
Collapse
Affiliation(s)
| | | | | | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32608, USA; (I.A.-P.); (A.C.); (S.D.)
| |
Collapse
|