1
|
Wang D, Zhang J, Bu C, Liu G, Guo G, Zhang Z, Lv G, Sheng Z, Yan Z, Gao Y, Wang M, Liu G, Zhao R, Li T, Ma C, Bu X. Dynamics of tumor in situ fluid circulating tumor DNA in recurrent glioblastomas forecasts treatment efficacy of immune checkpoint blockade coupled with low-dose bevacizumab. J Cancer Res Clin Oncol 2024; 150:466. [PMID: 39422764 PMCID: PMC11489198 DOI: 10.1007/s00432-024-05997-8] [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: 06/18/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
PURPOSE Immune checkpoint blockade (ICB) therapies have shown efficacy in various tumors, but long-term responses in glioblastoma are less than 10%. Quantifying tumor in situ fluid circulating tumor DNA (TISF-ctDNA) and therapeutic dynamics may enable real-time GBM disease burden evaluation. This study explores the potential of tumor in situ fluid circulating tumor DNA (TISF-ctDNA) dynamics in predicting treatment efficacy. METHODS TISF and peripheral blood samples were collected from patients with recurrent glioblastoma (rGBM) undergoing tislelizumab (a programmed death 1 inhibitor) combined with low-dose bevacizumab (an anti-vascular endothelial growth factor antibody) treatment before and during each immunotherapy cycle. Biomarkers evaluated included TISF-ctDNA, measured using Next Generation Sequencing (NGS), and host inflammation markers such as the platelet-to-lymphocyte ratio (PLR). RESULTS All 32 patients received tislelizumab plus low-dose bevacizumab regularly. The median progression-free survival (PFS) was 4.0 months, and overall survival (OS) was 22.3 months. An analysis of 19 patients with continuous evaluable TISF showed baseline TISF-ctDNA abundance did not correlate with OS (p = 0.23) or PFS (p = 0.23). However, a change in TISF-ctDNA maximal Somatic Variant Allelic Frequency (MVAF) after six treatment cycles predicted both PFS (p = 0.02) and OS (p < 0.0001). Lower baseline PLR also correlated with better survival outcomes. CONCLUSION The combination of tislelizumab and low-dose bevacizumab therapy appears to be effective in extending both OS and PFS in rGBM patients. Continuous TISF-ctDNA testing shows potential utility in complementing radiological monitoring. The temporal change pattern of TISF MVAF is more predictive of immunotherapy response than imaging. PLR before immunotherapy can screen patients likely to benefit from tislelizumab plus low-dose bevacizumab therapy. TRIAL REGISTRATION The trial registration number: NCT05502991; Date of registration: 2022-08-14.
Collapse
Affiliation(s)
- Dayang Wang
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Jiubing Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Chaojie Bu
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
- Department of Psychological Medicine, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, Henan, China
| | - Guanzheng Liu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Guangzhong Guo
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Ziyue Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Guangming Lv
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhiyuan Sheng
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhaoyue Yan
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Yvshuai Gao
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Gang Liu
- Department of Center for Clinical Single Cell Biomedicine, Department of Oncology, Clinical Research Center, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Ruijiao Zhao
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Tianxiao Li
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chunxiao Ma
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China
| | - Xingyao Bu
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China.
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China.
- Precision Diagnosis and Treatment Engineering Research Center for Glioma Henan Province, Zhengzhou, China.
- Clinical Center for Glioma Henan Provincial People's Hospital, Zhengzhou, China.
| |
Collapse
|
2
|
Yuan Y, Mishra F, Li B, Peng G, Chan P, Yang J, Liu Z. Modulating Tumor Immunity by Targeting Tumor Fibrotic Stroma and Angiogenic Vessels for Lung Cancer Treatment. Cancers (Basel) 2024; 16:2483. [PMID: 39001545 PMCID: PMC11240634 DOI: 10.3390/cancers16132483] [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: 06/02/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
Fibrotic stroma and angiogenic tumor vessels play an important role in modulating tumor immunity. We previously reported a rationally designed protein (ProAgio) that targets integrin αvβ3 at a novel site. ProAgio induces the apoptosis of cells that express high levels of the integrin. Both activated cancer-associated fibroblasts (CAFs) and angiogenic endothelial cells (aECs) in tumors express high levels of integrin αvβ3. ProAgio simultaneously and specifically induces apoptosis in CAFs and aECs in tumors. We provide evidence here that the depletion of CAFs and the elimination of leaky tumor angiogenic vessels by ProAgio alter tumor immunity. ProAgio reduces CD4+ Treg and Myeloid-derived suppressor cells (MDSCs), increases CD8+ T-cells, and increases the M1/M2 macrophage ratio in the tumor. The depletion of dense fibrotic stroma (CAFs) by ProAgio decreases the Programmed Death Ligand 1 (PDL-1) levels in the stroma areas surrounding the tumors, and thus strongly increases the delivery of anti-PDL-1 antibody to the target cancer cells. The impact of ProAgio on tumor immunity provides strong synergistical effects of checkpoint inhibitors on lung cancer treatment.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Falguni Mishra
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Bin Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Guangda Peng
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Payton Chan
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| | - Jenny Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA;
| | - Zhiren Liu
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (Y.Y.); (F.M.); (B.L.); (G.P.); (P.C.)
| |
Collapse
|
3
|
Argentiero A, Andriano A, Caradonna IC, de Martino G, Desantis V. Decoding the Intricate Landscape of Pancreatic Cancer: Insights into Tumor Biology, Microenvironment, and Therapeutic Interventions. Cancers (Basel) 2024; 16:2438. [PMID: 39001498 PMCID: PMC11240778 DOI: 10.3390/cancers16132438] [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: 05/30/2024] [Revised: 06/24/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) presents significant oncological challenges due to its aggressive nature and poor prognosis. The tumor microenvironment (TME) plays a critical role in progression and treatment resistance. Non-neoplastic cells, such as cancer-associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs), contribute to tumor growth, angiogenesis, and immune evasion. Although immune cells infiltrate TME, tumor cells evade immune responses by secreting chemokines and expressing immune checkpoint inhibitors (ICIs). Vascular components, like endothelial cells and pericytes, stimulate angiogenesis to support tumor growth, while adipocytes secrete factors that promote cell growth, invasion, and treatment resistance. Additionally, perineural invasion, a characteristic feature of PDAC, contributes to local recurrence and poor prognosis. Moreover, key signaling pathways including Kirsten rat sarcoma viral oncogene (KRAS), transforming growth factor beta (TGF-β), Notch, hypoxia-inducible factor (HIF), and Wnt/β-catenin drive tumor progression and resistance. Targeting the TME is crucial for developing effective therapies, including strategies like inhibiting CAFs, modulating immune response, disrupting angiogenesis, and blocking neural cell interactions. A recent multi-omic approach has identified signature genes associated with anoikis resistance, which could serve as prognostic biomarkers and targets for personalized therapy.
Collapse
Affiliation(s)
| | - Alessandro Andriano
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Ingrid Catalina Caradonna
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| | - Giulia de Martino
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70121 Bari, Italy
| | - Vanessa Desantis
- Department of Precision and Regenerative Medicine and Ionian Area, Pharmacology Section, Medical School, University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
4
|
Mo CC, Richardson E, Calabretta E, Corrado F, Kocoglu MH, Baron RM, Connors JM, Iacobelli M, Wei LJ, Rapoport AP, Díaz-Ricart M, Moraleda JM, Carlo-Stella C, Richardson PG. Endothelial injury and dysfunction with emerging immunotherapies in multiple myeloma, the impact of COVID-19, and endothelial protection with a focus on the evolving role of defibrotide. Blood Rev 2024; 66:101218. [PMID: 38852017 DOI: 10.1016/j.blre.2024.101218] [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: 05/01/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/10/2024]
Abstract
Patients with multiple myeloma (MM) were among the groups impacted more severely by the COVID-19 pandemic, with higher rates of severe disease and COVID-19-related mortality. MM and COVID-19, plus post-acute sequelae of SARS-CoV-2 infection, are associated with endothelial dysfunction and injury, with overlapping inflammatory pathways and coagulopathies. Existing treatment options for MM, notably high-dose therapy with autologous stem cell transplantation and novel chimeric antigen receptor (CAR) T-cell therapies and bispecific T-cell engaging antibodies, are also associated with endothelial cell injury and mechanism-related toxicities. These pathologies include cytokine release syndrome (CRS) and neurotoxicity that may be exacerbated by underlying endotheliopathies. In the context of these overlapping risks, prophylaxis and treatment approaches mitigating the inflammatory and pro-coagulant effects of endothelial injury are important considerations for patient management, including cytokine receptor antagonists, thromboprophylaxis with low-molecular-weight heparin and direct oral anticoagulants, and direct endothelial protection with defibrotide in the appropriate clinical settings.
Collapse
Affiliation(s)
- Clifton C Mo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA
| | - Edward Richardson
- Department of Medicine, Warren Alpert Medical School at Brown University, Providence, RI, USA
| | - Eleonora Calabretta
- Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Francesco Corrado
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA; Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy; Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Mehmet H Kocoglu
- Department of Medicine, University of Maryland School of Medicine, and Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Rebecca M Baron
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Lee-Jen Wei
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aaron P Rapoport
- Department of Medicine, University of Maryland School of Medicine, and Transplant and Cellular Therapy Program, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Maribel Díaz-Ricart
- Hematopathology, Pathology Department, CDB, Hospital Clinic, and IDIBAPS, Barcelona, Spain, and Barcelona Endothelium Team, Barcelona, Spain
| | - José M Moraleda
- Department of Medicine, Faculty of Medicine, Institute of Biomedical Research (IMIB-Pascual Parrilla), University of Murcia, Murcia, Spain
| | - Carmelo Carlo-Stella
- Department of Biomedical Sciences, Humanitas University, and IRCCS Humanitas Research Hospital, Milan, Italy
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Jerome Lipper Center for Multiple Myeloma Research, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Uziel O, Lipshtein L, Sarsor Z, Beery E, Bogen S, Lahav M, Regev A, Kliminski V, Sharan R, Gervits A, Signorini LF, Shimony S, Raanani P, Rozovski U. Chronic Lymphocytic Leukemia (CLL)-Derived Extracellular Vesicles Educate Endothelial Cells to Become IL-6-Producing, CLL-Supportive Cells. Biomedicines 2024; 12:1381. [PMID: 39061955 PMCID: PMC11273944 DOI: 10.3390/biomedicines12071381] [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: 05/08/2024] [Revised: 06/07/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
We hypothesized that via extracellular vesicles (EVs), chronic lymphocytic leukemia (CLL) cells turn endothelial cells into CLL-supportive cells. To test this, we treated vein-derived (HUVECs) and artery-derived (HAOECs) endothelial cells with EVs isolated from the peripheral blood of 45 treatment-naïve patients. Endothelial cells took up CLL-EVs in a dose- and time-dependent manner. To test whether CLL-EVs turn endothelial cells into IL-6-producing cells, we exposed them to CLL-EVs and found a 50% increase in IL-6 levels. Subsequently, we filtered out the endothelial cells and added CLL cells to this IL-6-enriched medium. After 15 min, STAT3 became phosphorylated, and there was a 40% decrease in apoptosis rate, indicating that IL-6 activated the STAT3-dependent anti-apoptotic pathway. Phospho-proteomics analysis of CLL-EV-exposed endothelial cells revealed 23 phospho-proteins that were upregulated, and network analysis unraveled the central role of phospho-β-catenin. We transfected HUVECs with a β-catenin-containing plasmid and found by ELISA a 30% increase in the levels of IL-6 in the culture medium. By chromatin immunoprecipitation assay, we observed an increased binding of three transcription factors to the IL-6 promoter. Importantly, patients with CLL possess significantly higher levels of peripheral blood IL-6 compared to normal individuals, suggesting that the inducers of endothelial IL-6 are the neoplastic EVs derived from the CLL cells versus those of healthy people. Taken together, we found that CLL cells communicate with endothelial cells through EVs that they release. Once they are taken up by endothelial cells, they turn them into IL-6-producing cells.
Collapse
Affiliation(s)
- Orit Uziel
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| | - Lian Lipshtein
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| | - Zinab Sarsor
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
| | - Einat Beery
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
| | - Shaked Bogen
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
| | - Meir Lahav
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| | - Alon Regev
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
| | - Vitali Kliminski
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
| | - Roded Sharan
- Blavatnik School of Computer Science, Tel-Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (R.S.); (A.G.); (L.F.S.)
| | - Asia Gervits
- Blavatnik School of Computer Science, Tel-Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (R.S.); (A.G.); (L.F.S.)
| | - Lorenzo Federico Signorini
- Blavatnik School of Computer Science, Tel-Aviv University, Ramat-Aviv, Tel Aviv 69978, Israel; (R.S.); (A.G.); (L.F.S.)
| | - Shai Shimony
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| | - Pia Raanani
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| | - Uri Rozovski
- The Felsenstein Medical Research Center, Rabin Medical Center Petah-Tikva, Petah Tikva 49100, Israel; (L.L.); (Z.S.); (E.B.); (M.L.); (A.R.); (V.K.); (S.S.); (P.R.); (U.R.)
- Institute of Hematology, Davidoff Cancer Center, Petah Tikva 49100, Israel;
- Faculty of Medicine, Tel-Aviv University, Ramat-Aviv, Tel Aviv 6997801, Israel
| |
Collapse
|
6
|
Tojjari A, Park R, Yu J, Saeed A. Targeting Angiogenesis Alone and in Combination with Immune Checkpoint Inhibitors in Advanced Gastroesophageal Malignancies. Curr Gastroenterol Rep 2024; 26:57-67. [PMID: 38294661 DOI: 10.1007/s11894-024-00920-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW This review critically examines the latest approaches in treating advanced gastroesophageal malignancies. It emphasizes the significance of angiogenesis as a therapeutic target and discusses the potential synergy of combining angiogenesis inhibitors with immune checkpoint inhibitors (ICIs) to enhance treatment efficacy. RECENT FINDINGS Emerging evidence from clinical trials, such as the INTEGRATE IIa trial with regorafenib and studies involving apatinib and sunitinib, underscores the efficacy of targeting the VEGFR pathway. These studies indicate substantial benefits in progression-free survival (PFS) and overall survival (OS) in patients with advanced stages of the disease who have limited treatment options. Additionally, the recent introduction of combination therapies involving ICIs has shown an increased response rate, suggesting a promising direction for future treatment protocols. The landscape of treatment for gastroesophageal malignancies is rapidly evolving. Research is now pivoting from conventional chemotherapy to a more nuanced approach that includes targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Alireza Tojjari
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 15213, USA
| | - Robin Park
- Division of Hematology and Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - James Yu
- Division of Hematology and Medical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, 15213, USA.
| |
Collapse
|
7
|
Yue M, Chen MM, Zhang B, Wang Y, Li P, Zhao Y. The functional roles of chemokines and chemokine receptors in colorectal cancer progression. Biomed Pharmacother 2024; 170:116040. [PMID: 38113624 DOI: 10.1016/j.biopha.2023.116040] [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: 09/27/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023] Open
Abstract
Colorectal cancer is a common malignancy with significant rates of morbidity and mortality. A number of factors, including the tumor microenvironment, chemokines, the inflammatory response, have an impact on the development of colorectal cancer. A critical component of the tumor microenvironment is chemokines. Various cell subsets are attracted to the tumor microenvironment through interactions with chemokine receptors. These cells have varying effects on the development of the tumor and the effectiveness of treatment. Additionally, chemokines can participate in inflammatory processes and have effects that are either pro- or anti-tumor. Chemokines can be exploited as targets for medication resistance and treatment in colorectal cancer. In this review, we discuss the expression of chemokines and chemokine receptors, and their relationship with immune cells in the tumor microenvironment. At the same time, we also collect and discuss the significance of chemokines and chemokine receptors in colorectal cancer progression, and their potential as molecular targets for CRC treatment.
Collapse
Affiliation(s)
- Mingli Yue
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Meng-Meng Chen
- Key Laboratory of Cancer and Immune Cells of Qingdao, Qingdao 266021, China; Qingdao Restore Biotechnology Co., Ltd., Qingdao, Shandong 266111, PR China
| | - Bingqiang Zhang
- Key Laboratory of Cancer and Immune Cells of Qingdao, Qingdao 266021, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yi Zhao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, China; Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Maternal and Child Health Care Hospital of Shandong Province affiliated to Qingdao University, Shandong Province, China.
| |
Collapse
|
8
|
Jariani A, Kakroodi ST, Arabfard M, Jamialahmadi T, Rahimi M, Sahebkar A. Identification of Key Genes in Angiogenesis of Breast and Prostate Cancers in the Context of Different Cell Types. Curr Med Chem 2024; 31:1595-1605. [PMID: 36999716 DOI: 10.2174/0929867330666230331101458] [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: 10/13/2022] [Revised: 12/27/2022] [Accepted: 02/03/2023] [Indexed: 04/01/2023]
Abstract
INTRODUCTION Angiogenesis involves the development of new blood vessels. Biochemical signals start this process in the body, which is followed by migration, growth, and differentiation of endothelial cells that line the inside wall of blood vessels. This process is vital for the growth of cancer cells and tumors. MATERIALS AND METHODS We started our analysis by composing a list of genes that have a validated impact in humans with respect to angiogenesis-related phenotypes. Here, we have investigated the expression patterns of angiogenesis-related genes in the context of previously published single-cell RNA-Seq data from prostate and breast cancer samples. RESULTS Using a protein-protein interaction network, we showed how different modules of angiogenesis-related genes are overexpressed in different cell types. In our results, genes, such as ACKR1, AQP1, and EGR1, showed a strong cell type-dependent overexpression pattern in the two investigated cancer types, which can potentially be helpful in the diagnosis and follow-up of patients with prostate and breast cancer. CONCLUSION Our work demonstrates how different biological processes in distinct cell types contribute to the angiogenesis process, which can provide clues regarding the potential application of targeted inhibition of the angiogenesis process.
Collapse
Affiliation(s)
- Abbas Jariani
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Masoud Arabfard
- Chemical Injuries Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Tannaz Jamialahmadi
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rahimi
- Clinical care and Health Promotion Research Center, Karaj Branch, Islamic Azad University, Karaj, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Schirizzi A, Arshadi A, Tolomeo D, Schirosi L, Valentini AM, De Leonardis G, Refolo MG, Donghia R, Storlazzi CT, Zito A, Ricci AD, Vallarelli S, Ostuni C, Bencivenga M, De Manzoni G, Messa C, Armentano R, Giannelli G, Lotesoriere C, D’Alessandro R. VEGFA Status as a Predictive Marker of Therapy Outcome in Metastatic Gastric Cancer Patients Following Ramucirumab-Based Treatment. Biomedicines 2023; 11:2721. [PMID: 37893095 PMCID: PMC10603940 DOI: 10.3390/biomedicines11102721] [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: 09/06/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
Metastatic gastric cancer (mGC) often has a poor prognosis and may benefit from a few targeted therapies. Ramucirumab-based anti-angiogenic therapy targeting the VEGFR2 represents a milestone in the second-line treatment of mGC. Several studies on different cancers are focusing on the major VEGFR2 ligand status, meaning VEGFA gene copy number and protein overexpression, as a prognostic marker and predictor of response to anti-angiogenic therapy. Following this insight, our study aims to examine the role of VEGFA status as a predictive biomarker for the outcome of second-line therapy with Ramucirumab and paclitaxel in mGC patients. To this purpose, the copy number of the VEGFA gene, by fluorescence in situ hybridization experiments, and its expression in tumor tissue as well as the density of micro-vessels, by immunohistochemistry experiments, were assessed in samples derived from mGC patients. This analysis found that amplification of VEGFA concomitantly with VEGFA overexpression and overexpression of VEGFA with micro-vessels density are more represented in patients showing disease control during treatment with Ramucirumab. In addition, in the analyzed series, it was found that amplification was not always associated with overexpression of VEGFA, but overexpression of VEGFA correlates with high micro-vessel density. In conclusion, overexpression of VEGFA could emerge as a potential biomarker to predict the response to anti-angiogenic therapy.
Collapse
Affiliation(s)
- Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Aram Arshadi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Doron Tolomeo
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Laura Schirosi
- Pathology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (L.S.); (A.Z.)
| | - Anna Maria Valentini
- Histopathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.M.V.); (R.A.)
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Maria Grazia Refolo
- Clinical Pathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Rossella Donghia
- Data Science Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Clelia Tiziana Storlazzi
- Department of Biosciences, Biotechnology and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (A.A.); (D.T.); (C.T.S.)
| | - Alfredo Zito
- Pathology Department, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (L.S.); (A.Z.)
| | - Angela Dalia Ricci
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Simona Vallarelli
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Carmela Ostuni
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Maria Bencivenga
- General and Upper GI Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Giovanni De Manzoni
- General and Upper GI Surgery Division, University of Verona, 37126 Verona, Italy; (M.B.); (G.D.M.)
| | - Caterina Messa
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| | - Raffaele Armentano
- Histopathology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.M.V.); (R.A.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy;
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.D.R.); (S.V.); (C.O.)
| | - Rosalba D’Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (A.S.); (G.D.L.); (C.M.)
| |
Collapse
|
10
|
Wang S, Wang Y, Zhu H, Chen M, Zhang L. Gambogic Acid Inhibits Gastric Cancer Cell Proliferation through Necroptosis. Can J Gastroenterol Hepatol 2023; 2023:7532367. [PMID: 37588664 PMCID: PMC10427235 DOI: 10.1155/2023/7532367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 06/06/2023] [Accepted: 07/05/2023] [Indexed: 08/18/2023] Open
Abstract
Gambogic acid (GA) is a natural xanthonoid secreted by Garcinia hanburyi tree. It possesses anti-cancer activity in various types of cancers. In gastric cancer, it inhibits cell proliferation through increasing apoptosis. However, whether necroptosis is involved in the GA-induced proliferation inhibited in gastric cancer is unknown. In the present study, we found that RIPK1 specific inhibitor necrostatin-1 (Nec-1) attenuated GA-induced proliferation inhibition. GA treatment increased the phosphorylation of necroptosis-related proteins, RIPK1, RIPK3, and MLKL, and their interactions to form the necrosome complex. The effector protein Drp-1 was dephosphorylated by GA treatment. Inhibition of necroptosis by different inhibitors and PGAM5 knockdown attenuated GA-induced cell death in gastric cancer cell lines, thereby attenuating GA-caused cell proliferation inhibition. All the data supported the conclusion that GA could inhibit gastric cancer cell proliferation by inducing necroptosis.
Collapse
Affiliation(s)
- Shujun Wang
- Department of Gastroenterology, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Yiping Wang
- Department of Gastroenterology, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Hui Zhu
- Department of Gastroenterology, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Miaohui Chen
- Department of Gastroenterology, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| | - Liang Zhang
- Department of Gastroenterology, Affiliated Cixi People's Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315300, China
| |
Collapse
|
11
|
Katsi V, Papakonstantinou I, Tsioufis K. Atherosclerosis, Diabetes Mellitus, and Cancer: Common Epidemiology, Shared Mechanisms, and Future Management. Int J Mol Sci 2023; 24:11786. [PMID: 37511551 PMCID: PMC10381022 DOI: 10.3390/ijms241411786] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The involvement of cardiovascular disease in cancer onset and development represents a contemporary interest in basic science. It has been recognized, from the most recent research, that metabolic syndrome-related conditions, ranging from atherosclerosis to diabetes, elicit many pathways regulating lipid metabolism and lipid signaling that are also linked to the same framework of multiple potential mechanisms for inducing cancer. Otherwise, dyslipidemia and endothelial cell dysfunction in atherosclerosis may present common or even interdependent changes, similar to oncogenic molecules elevated in many forms of cancer. However, whether endothelial cell dysfunction in atherosclerotic disease provides signals that promote the pre-clinical onset and proliferation of malignant cells is an issue that requires further understanding, even though more questions are presented with every answer. Here, we highlight the molecular mechanisms that point to a causal link between lipid metabolism and glucose homeostasis in metabolic syndrome-related atherosclerotic disease with the development of cancer. The knowledge of these breakthrough mechanisms may pave the way for the application of new therapeutic targets and for implementing interventions in clinical practice.
Collapse
Affiliation(s)
- Vasiliki Katsi
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
| | | | - Konstantinos Tsioufis
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
12
|
Thomas JA, Gireesh Moly AG, Xavier H, Suboj P, Ladha A, Gupta G, Singh SK, Palit P, Babykutty S. Enhancement of immune surveillance in breast cancer by targeting hypoxic tumor endothelium: Can it be an immunological switch point? Front Oncol 2023; 13:1063051. [PMID: 37056346 PMCID: PMC10088512 DOI: 10.3389/fonc.2023.1063051] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/17/2023] [Indexed: 03/30/2023] Open
Abstract
Breast cancer ranks second among the causes of cancer-related deaths in women. In spite of the recent advances achieved in the diagnosis and treatment of breast cancer, further study is required to overcome the risk of cancer resistance to treatment and thereby improve the prognosis of individuals with advanced-stage breast cancer. The existence of a hypoxic microenvironment is a well-known event in the development of mutagenesis and rapid proliferation of cancer cells. Tumor cells, purposefully cause local hypoxia in order to induce angiogenesis and growth factors that promote tumor growth and metastatic characteristics, while healthy tissue surrounding the tumor suffers damage or mutate. It has been found that these settings with low oxygen levels cause immunosuppression and a lack of immune surveillance by reducing the activation and recruitment of tumor infiltrating leukocytes (TILs). The immune system is further suppressed by hypoxic tumor endothelium through a variety of ways, which creates an immunosuppressive milieu in the tumor microenvironment. Non responsiveness of tumor endothelium to inflammatory signals or endothelial anergy exclude effector T cells from the tumor milieu. Expression of endothelial specific antigens and immunoinhibitory molecules like Programmed death ligand 1,2 (PDL-1, 2) and T cell immunoglobulin and mucin-domain containing-3 (TIM-3) by tumor endothelium adds fuel to the fire by inhibiting T lymphocytes while promoting regulatory T cells. The hypoxic microenvironment in turn recruits Myeloid Derived Suppressor Cells (MDSCs), Tumor Associated Macrophages (TAMs) and T regulatory cells (Treg). The structure and function of newly generated blood vessels within tumors, on the other hand, are aberrant, lacking the specific organization of normal tissue vasculature. Vascular normalisation may work for a variety of tumour types and show to be an advantageous complement to immunotherapy for improving tumour access. By enhancing immune response in the hypoxic tumor microenvironment, via immune-herbal therapeutic and immune-nutraceuticals based approaches that leverage immunological evasion of tumor, will be briefly reviewed in this article. Whether these tactics may be the game changer for emerging immunological switch point to attenuate the breast cancer growth and prevent metastatic cell division, is the key concern of the current study.
Collapse
Affiliation(s)
- Juvin Ann Thomas
- Centre for Tumor Immunology and Microenvironment, Department of Zoology, Mar Ivanios College, Nalanchira, Thiruvananthapuram, Kerala, India
| | - Athira Gireesh Gireesh Moly
- Centre for Tumor Immunology and Microenvironment, Department of Zoology, Mar Ivanios College, Nalanchira, Thiruvananthapuram, Kerala, India
| | - Hima Xavier
- Centre for Tumor Immunology and Microenvironment, Department of Zoology, Mar Ivanios College, Nalanchira, Thiruvananthapuram, Kerala, India
| | - Priya Suboj
- Department of Botany and Biotechnology, St. Xaviers College, Thumba, Thiruvananthapuram, Kerala, India
| | - Amit Ladha
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, West-Midlands, United Kingdom
| | - Gaurav Gupta
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Santosh Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Partha Palit
- Drug Discovery Research Laboratory, Assam University, Silchar, Department of Pharmaceutical Sciences, Assam, India
| | - Suboj Babykutty
- Centre for Tumor Immunology and Microenvironment, Department of Zoology, Mar Ivanios College, Nalanchira, Thiruvananthapuram, Kerala, India
| |
Collapse
|
13
|
Zhao X, Sakamoto S, Wei J, Pae S, Saito S, Sazuka T, Imamura Y, Anzai N, Ichikawa T. Contribution of the L-Type Amino Acid Transporter Family in the Diagnosis and Treatment of Prostate Cancer. Int J Mol Sci 2023; 24:ijms24076178. [PMID: 37047148 PMCID: PMC10094571 DOI: 10.3390/ijms24076178] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
The L-type amino acid transporter (LAT) family contains four members, LAT1~4, which are important amino acid transporters. They mainly transport specific amino acids through cell membranes, provide nutrients to cells, and are involved in a variety of metabolic pathways. They regulate the mTOR signaling pathway which has been found to be strongly linked to cancer in recent years. However, in the field of prostate cancer (PCa), the LAT family is still in the nascent stage of research, and the importance of LATs in the diagnosis and treatment of prostate cancer is still unknown. Therefore, this article aims to report the role of LATs in prostate cancer and their clinical significance and application. LATs promote the progression of prostate cancer by increasing amino acid uptake, activating the mammalian target of rapamycin (mTOR) pathway and downstream signals, mediating castration-resistance, promoting tumor angiogenesis, and enhancing chemotherapy resistance. The importance of LATs as diagnostic and therapeutic targets for prostate cancer was emphasized and the latest research results were introduced. In addition, we introduced selective LAT1 inhibitors, including JPH203 and OKY034, which showed excellent inhibitory effects on the proliferation of various tumor cells. This is the future direction of amino acid transporter targeting therapy drugs.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Jiaxing Wei
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Sangjon Pae
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Shinpei Saito
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomokazu Sazuka
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Yusuke Imamura
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Ichikawa
- Department of Urology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
14
|
Kang J, Xiang X, Chen X, Jiang J, Zhang Y, Li L, Tang J. Angiogenesis-related gene signatures reveal the prognosis of cervical cancer based on single cell sequencing and co-expression network analysis. Front Cell Dev Biol 2023; 10:1086835. [PMID: 36712973 PMCID: PMC9877352 DOI: 10.3389/fcell.2022.1086835] [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: 11/01/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
Cervical cancer ranks first in female reproductive tract tumors in terms of morbidity and mortality. Yet the curative effect of patients with persistent, recurrent or metastatic cervical cancer remains unsatisfactory. Although antitumor angiogenic drugs have been recommended as the first-line treatment options for cervical cancer, there are no comprehensive prognostic indicators for cervical cancer based on angiogenic signature genes. In this study, we aimed to develop a model to assess the prognosis of cervical cancer based on angiogenesis-related (AG) signature genes, and to provide some reference for the comprehensive treatment of cervical cancer in the clinical setting. First we screened the AG gene set from GeneCard website, and then performed angiogenesis-related scores (AGS) per cell from single cell sequencing dataset GSE168652, followed by performing weighted gene co-expression network analysis (WGCNA) for cervical cancer patients according to angiogenesis phenotype. Thus, we established a prognostic model based on AGS by taking the intersection of WGCNA angiogenic module gene and differential gene (DEGs) of GSE168652. The GSE44001 was selected as an external validation set, followed by performing ROC curve analysis to assess its accuracy. The results showed that we successfully constructed a prognostic model related to the AG genes. Patients in the high-AGS group in both the train, test and the validation sets had a worse prognosis than those in the low-AGS group, had lower expression of most immune checkpoint-associated genes and lower tumor mutational burden as well. Patients in the low-AGS group were more sensitive to AMG.706, Bosutinib, and Lenalidomide while Imatinib, Pazopanib, and Sorafenib were more recommended to patients in the high-AGS group. Finally, TXNDC12 and ZC3H13, which have high hazard ratio and poor prognosis in the model, were highly expressed in cervical cancer cell lines and tissue. Meanwhile, the results showed that TXNDC12 promoted the migration of cervical cancer cells and the tubule-forming ability of endothelial cells. In conclusion, our model based on genes with AG features can effectively assess the prognosis of cervical cancer, and can also provide reference for clinicians to choose immune-related treatments.
Collapse
Affiliation(s)
- Jiawen Kang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China
| | - Xiaoqing Xiang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China
| | - Xiaoyan Chen
- Department of Pathology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jingwen Jiang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China
| | - Yong Zhang
- Department of Internal Medicine, Medical College of Hunan Normal University, Changsha, Hunan, China,*Correspondence: Yong Zhang, ; Lesai Li, ; Jie Tang,
| | - Lesai Li
- Department of Gynecologic Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China,*Correspondence: Yong Zhang, ; Lesai Li, ; Jie Tang,
| | - Jie Tang
- Department of Gynecologic Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China,*Correspondence: Yong Zhang, ; Lesai Li, ; Jie Tang,
| |
Collapse
|
15
|
Solimando AG, Desantis V, Da Vià MC. Visualizing the Interactions Shaping the Imaging of the Microenvironment in Human Cancers. Methods Mol Biol 2023; 2572:67-79. [PMID: 36161408 DOI: 10.1007/978-1-0716-2703-7_5] [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: 06/16/2023]
Abstract
The Visium Spatial Gene Expression Solution (Visium 10×) allows for the mRNA analysis using high throughput sequencing and maps a transcriptional expression pattern in tissue sections using high-resolution microscope imaging in ex-vivo human and mice samples. The workflow surveys spatial global gene expression in tissue sections, exploiting the whole transcriptome profiling and defining the set of transcripts via targeted gene panels. An automated cell type annotation allows a comparison with control tissue samples. This technique delineates cancerous or diseased tissue boundaries and details gene expression gradients in the tissue surrounding the tumor or pathologic nests. Remarkably, the Visium 10× allows for whole transcriptome and targeted analysis without the loss of spatial information. This approach provides gene expression data within the context of tissue architecture, tissue microenvironments, and cell groups. It can be used in association with therapy, anti-angiogenic therapy, and immunotherapy to improve treatment response.
Collapse
Affiliation(s)
- Antonio G Solimando
- Department of Biomedical Sciences and Human Oncology (DIMO), Unit of Internal Medicine and Clinical Oncology, University of Bari Aldo Moro Medical School, Bari, Italy.
| | - Vanessa Desantis
- Department of Biomedical Sciences and Human Oncology (DIMO), Pharmacology Section, Medical School, University of Bari Aldo Moro, Bari, Italy
| | - Matteo Claudio Da Vià
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
16
|
The Prognostic Significance of FKBP1A and Its Related Immune Infiltration in Liver Hepatocellular Carcinoma. Int J Mol Sci 2022; 23:ijms232112797. [PMID: 36361587 PMCID: PMC9659304 DOI: 10.3390/ijms232112797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/06/2022] [Accepted: 10/18/2022] [Indexed: 12/24/2022] Open
Abstract
Liver hepatocellular carcinoma (LIHC) remains a global health challenge with poor prognosis and high mortality. FKBP1A was first discovered as a receptor for the immunosuppressant drug FK506 in immune cells and is critical for various tumors and cancers. However, the relationships between FKBP1A expression, cellular distribution, tumor immunity, and prognosis in LIHC remain unclear. Here, we investigated the expression level of FKBP1A and its prognostic value in LIHC via multiple datasets including ONCOMINE, TIMER, GEPIA, UALCAN, HCCDB, Kaplan–Meier plotter, LinkedOmics, and STRING. Human liver tissue microarray was employed to analyze the characteristics of FKBP1A protein including the expression level and pathological alteration in cellular distribution. FKBP1A expression was significantly higher in LIHC and correlated with tumor stage, grade and metastasis. The expression level of the FKBP1A protein was also increased in LIHC patients along with its accumulation in endoplasmic reticulum (ER). High FKBP1A expression was correlated with a poor survival rate in LIHC patients. The analysis of gene co-expression and the regulatory pathway network suggested that FKBP1A is mainly involved in protein synthesis, metabolism and the immune-related pathway. FKBP1A expression had a significantly positive association with the infiltration of hematopoietic immune cells including B cells, CD8+ T cells, CD4+ T cells, macrophages, neutrophils, and dendritic cells. Moreover, M2 macrophage infiltration was especially associated with a poor survival prognosis in LIHC. Furthermore, FKBP1A expression was significantly positively correlated with the expression of markers of M2 macrophages and immune checkpoint proteins such as PD-L1, CTLA-4, LAG3 and HAVCR2. Our study demonstrated that FKBP1A could be a potential prognostic target involved in tumor immune cell infiltration in LIHC.
Collapse
|
17
|
Maiorano BA, Parisi A, Maiello E, Ciardiello D. The Interplay between Anti-Angiogenics and Immunotherapy in Colorectal Cancer. LIFE (BASEL, SWITZERLAND) 2022; 12:life12101552. [PMID: 36294987 PMCID: PMC9604892 DOI: 10.3390/life12101552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022]
Abstract
Simple Summary Colorectal cancer is a frequent and lethal neoplasm. The tumor often creates new vessels to grow and spread—a process called ‘angiogenesis’. Therefore, drugs blocking angiogenesis are effective against this malignancy. On the other side, immune checkpoint inhibitors, which unleash the immune system to fight against tumors, have limited efficacy in patients carrying instability of DNA regions called microsatellites. However, there is an interaction between angiogenic factors and the immune system. This gives a chance to combine anti-angiogenic agents and immune checkpoint inhibitors to improve the efficacy of treating this malignancy. Abstract Angiogenesis, a hallmark of cancer, plays a fundamental role in colorectal cancer (CRC). Anti-angiogenic drugs and chemotherapy represent a standard of care for treating metastatic disease. Immune checkpoint inhibitors (ICIs) have changed the therapeutic algorithm of many solid tumors. However, the efficacy of ICIs is limited to mCRC patients carrying microsatellite instability (MSI-H), which represent approximately 3–5% of mCRC. Emerging evidence suggests that anti-angiogenic drugs could exhibit immunomodulatory properties. Thus, there is a strong rationale for combining anti-angiogenics and ICIs to improve efficacy in the metastatic setting. Our review summarizes the pre-clinical and clinical evidence regarding the combination of anti-angiogenics and ICIs in mCRC to deepen the possible application in daily clinical practice.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
- Correspondence:
| | - Alessandro Parisi
- Department of Oncology, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, 60126 Ancona, Italy
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Evaristo Maiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Davide Ciardiello
- Oncology Unit, IRCCS Foundation Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Medical Oncology Unit, Department of Precision Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| |
Collapse
|
18
|
Solimando AG, Malerba E, Leone P, Prete M, Terragna C, Cavo M, Racanelli V. Drug resistance in multiple myeloma: Soldiers and weapons in the bone marrow niche. Front Oncol 2022; 12:973836. [PMID: 36212502 PMCID: PMC9533079 DOI: 10.3389/fonc.2022.973836] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple myeloma (MM) is still an incurable disease, despite considerable improvements in treatment strategies, as resistance to most currently available agents is not uncommon. In this study, data on drug resistance in MM were analyzed and led to the following conclusions: resistance occurs via intrinsic and extrinsic mechanisms, including intraclonal heterogeneity, drug efflux pumps, alterations of drug targets, the inhibition of apoptosis, increased DNA repair and interactions with the bone marrow (BM) microenvironment, cell adhesion, and the release of soluble factors. Since MM involves the BM, interactions in the MM-BM microenvironment were examined as well, with a focus on the cross-talk between BM stromal cells (BMSCs), adipocytes, osteoclasts, osteoblasts, endothelial cells, and immune cells. Given the complex mechanisms that drive MM, next-generation treatment strategies that avoid drug resistance must target both the neoplastic clone and its non-malignant environment. Possible approaches based on recent evidence include: (i) proteasome and histone deacetylases inhibitors that not only target MM but also act on BMSCs and osteoclasts; (ii) novel peptide drug conjugates that target both the MM malignant clone and angiogenesis to unleash an effective anti-MM immune response. Finally, the role of cancer stem cells in MM is unknown but given their roles in the development of solid and hematological malignancies, cancer relapse, and drug resistance, their identification and description are of paramount importance for MM management.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- Istituto di ricovero e cura a carattere scientifico (IRCCS) Istituto Tumori ‘Giovanni Paolo II’ of Bari, Bari, Italy
| | - Eleonora Malerba
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Marcella Prete
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
| | - Carolina Terragna
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Michele Cavo
- ’Seràgnoli’ Institute of Hematology, Bologna University School of Medicine, Bologna, Italy
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, School of Medicine, ‘Aldo Moro’ University of Bari, Bari, Italy
- *Correspondence: Vito Racanelli,
| |
Collapse
|
19
|
Solimando AG, Marziliano D, Ribatti D. SARS-CoV-2 and Endothelial Cells: Vascular Changes, Intussusceptive Microvascular Growth and Novel Therapeutic Windows. Biomedicines 2022; 10:2242. [PMID: 36140343 PMCID: PMC9496230 DOI: 10.3390/biomedicines10092242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
Endothelial activation in infectious diseases plays a crucial role in understanding and predicting the outcomes and future treatments of several clinical conditions. COVID-19 is no exception. Moving from basic principles to novel approaches, an evolving view of endothelial activation provides insights into a better knowledge of the upstream actors in COVID-19 as a crucial future direction for managing SARS-CoV-2 and other infections. Assessing the function of resting and damaged endothelial cells in infection, particularly in COVID-19, five critical processes emerged controlling thrombo-resistance: vascular integrity, blood flow regulation, immune cell trafficking, angiogenesis and intussusceptive microvascular growth. Endothelial cell injury is associated with thrombosis, increased vessel contraction and a crucial phenomenon identified as intussusceptive microvascular growth, an unprecedented event of vessel splitting into two lumens through the integration of circulating pro-angiogenic cells. An essential awareness of endothelial cells and their phenotypic changes in COVID-19 inflammation is pivotal to understanding the vascular biology of infections and may offer crucial new therapeutic windows.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
| | - Donatello Marziliano
- Guido Baccelli Unit of Internal Medicine, Department of Biomedical Sciences and Human Oncology, School of Medicine, Aldo Moro University of Bari, 70124 Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
20
|
Hervás-Salcedo R, Martín-Antonio B. A Journey through the Inter-Cellular Interactions in the Bone Marrow in Multiple Myeloma: Implications for the Next Generation of Treatments. Cancers (Basel) 2022; 14:3796. [PMID: 35954459 PMCID: PMC9367481 DOI: 10.3390/cancers14153796] [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] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 02/05/2023] Open
Abstract
Tumors are composed of a plethora of extracellular matrix, tumor and non-tumor cells that form a tumor microenvironment (TME) that nurtures the tumor cells and creates a favorable environment where tumor cells grow and proliferate. In multiple myeloma (MM), the TME is the bone marrow (BM). Non-tumor cells can belong either to the non-hematological compartment that secretes soluble mediators to create a favorable environment for MM cells to grow, or to the immune cell compartment that perform an anti-MM activity in healthy conditions. Indeed, marrow-infiltrating lymphocytes (MILs) are associated with a good prognosis in MM patients and have served as the basis for developing different immunotherapy strategies. However, MM cells and other cells in the BM can polarize their phenotype and activity, creating an immunosuppressive environment where immune cells do not perform their cytotoxic activity properly, promoting tumor progression. Understanding cell-cell interactions in the BM and their impact on MM proliferation and the performance of tumor surveillance will help in designing efficient anti-MM therapies. Here, we take a journey through the BM, describing the interactions of MM cells with cells of the non-hematological and hematological compartment to highlight their impact on MM progression and the development of novel MM treatments.
Collapse
Affiliation(s)
| | - Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), University Autonomous of Madrid (UAM), 28040 Madrid, Spain
| |
Collapse
|
21
|
Albumin Paclitaxel Compared with 5-Penfluorouracil, Lobaplatin, and Albumin Paclitaxel Combined with 5-Penfluorouracil in the Treatment of Human Gastric Cancer Cell AGS Line Autophagy and Apoptosis. Can J Gastroenterol Hepatol 2022; 2022:6015877. [PMID: 35719323 PMCID: PMC9205742 DOI: 10.1155/2022/6015877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/14/2022] [Accepted: 04/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Gastric cancer is one of the most common malignant tumors in the world. Albumin paclitaxel (Nab-PTX) is a novel microtubule inhibitor with albumin as the carrier. Several clinical trials are underway in gastric cancer, but the autophagy mechanism of Nab-PTX on gastric cancer is still unclear. The autophagy and apoptosis effects of Nab-PTX compared with 5-pentafluorouracil (5-Fu) and lobaplatin (LBP) in gastric cancer are also unclear. OBJECTIVE This article will compare the effects of Nab-PTX, 5-Fu, LBP, and albumin paclitaxel + 5-pentafluorouracil (Nab-PTX + 5-Fu) on AGS cells from the perspective of autophagy and apoptosis, which is to provide new ideas and experimental evidence for gastric cancer. METHOD (1) Experimental groups were control (Ctrl), Nab-PTX, 5-Fu, LBP, and Nab-PTX + 5-Fu. (2) CCK-8 assay was used to reflect cell viability and proliferation. (3) The flow cytometry was used to perform the 24-hour apoptosis and cell cycle of each group. (4) Western blot assay was used to investigate autophagy signal proteins LC3I/LC3II, LC3II/LC3I, SQSTM1/p62, Beclin-1, Atg12, Atg5, p-mULK1, p-AMPK, p-mTOR, and apoptosis signal proteins Bax and Bcl-2. RESULTS Nab-PTX, 5-Fu, LBP, and Nab-PTX + 5-Fu inhibited AGS cells' proliferation and arrested the cell cycle. At the same time, each group increased the apoptosis of AGS cells to various degrees (Nab-PTX + 5-Fu > Nab-PTX > 5-Fu > LBP, respectively). The experimental results showed that Nab-PTX and Nab-PTX + 5-Fu promoted autophagy and apoptosis of AGS cells. The comparison of Nab-PTX, 5-Fu, and LBP between groups revealed that 5-Fu inhibited autophagy and the expression of apoptosis protein Bax. In LBP, abnormal activation of autophagy downstream, blocking of autophagy flow, abnormal increase of ATG12, and increased expression of apoptosis protein Bax occurred. Further study found that the autophagy upstream mechanism is different. CONCLUSION Nab-PTX, 5-Fu, LBP, and Nab-PTX + 5-Fu can inhibit cell proliferation, promote cell apoptosis, and induce the difference in autophagy expression. The autophagy difference of this antitumor drug may be related to its inducing apoptosis. Meanwhile, Nab-PTX has a better antitumor effect than 5-Fu and LBP in gastric cancer, and the combination of Nab-PTX + 5-Fu has more antitumor advantages.
Collapse
|
22
|
Shibata C, Nakano T, Yasumoto A, Mitamura A, Sawada K, Ogawa H, Miura T, Ise I, Takami K, Yamamoto K, Katayose Y. Comparison of CEA and CA19-9 as a predictive factor for recurrence after curative gastrectomy in gastric cancer. BMC Surg 2022; 22:213. [PMID: 35655198 PMCID: PMC9164336 DOI: 10.1186/s12893-022-01667-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 05/25/2022] [Indexed: 12/27/2022] Open
Abstract
Background Our aim of was to compare importance of the tumor markers (TMs) serum carcinoembryonic antigen (CEA) and carbohydrate antigen (CA) 19-9 in prediction of recurrence after curative gastrectomy for gastric cancer. Methods We reviewed retrospectively the clinical records of 149 patients who underwent curative gastrectomy for stage I–III gastric cancer and whose CEA and CA19-9 levels were determined once preoperatively and for more than 3 years postoperatively. We investigated whether the clinicopathological characteristics of patients including age, sex, pathological disease stage, operative approach, type of gastrectomy, and degree of lymph node dissection as well as preoperative positivity of CEA and CA19-9 were risk factors for recurrence in univariate and multivariate analyses. Rate of recurrence was compared between patients positive and negative for postoperative CEA or CA19-9. We also calculated sensitivity, specificity, positive and negative predictable values of postoperative positivity of CEA and CA19-9 for recurrence. The lead time was compared between CEA and CA19-9 that was defined as the time of the first detection of increases in tumor markers and confirmation of recurrence on imaging modalities. Results The number of patients positive for preoperative CEA was 25 (17%) and for CA19-9 was 11 (7%). Recurrence was confirmed in 29 (19%) patients. Stage III disease, preoperative positivity for CA19-9 but not CEA, and total gastrectomy were risk factors for recurrence in univariate analysis, but stage III disease was the only risk factor for recurrence in multivariate analysis. Forty and 15 patients were positive for postoperative CEA and CA19-9, respectively. The recurrence rate of 47% (7/15) in patients positive for postoperative CA19-9 was greater than that in negative patients (22/134 = 16%), but it did not differ between patients who were positive or negative for postoperative CEA. Specificity for CA19-9 was greater than that for CEA (P < 0.05). The lead time of CEA (3.9 ± 4.7 months) was not different from that of CA19-9 (6.1 ± 7.1 months). Conclusions These results indicate that CA19-9 rather than CEA is likely to be more useful for the detection of recurrence after curative gastrectomy for gastric cancer.
Collapse
|
23
|
Xu Y, Xiong Q, Yang Y, Weng N, Li J, Liu J, Yang X, Zeng Z, Zhang Z, Zhu Q. Serum Nardilysin as a Prognostic Biomarker in Pancreatic Ductal Adenocarcinoma. J Clin Med 2022; 11:jcm11113101. [PMID: 35683488 PMCID: PMC9181681 DOI: 10.3390/jcm11113101] [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: 04/18/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Nardilysin, (N-arginine dibasic convertase, NRDC) has been reported to play an important role in cancer progression, and is associated with tumor proliferation signals and inflammatory signals, such as tumor necrosis factor-a (TNF-a) and heparin-binding epidermal growth factor-like growth factor (HB-EGF), through the activation of disintegrin and metalloproteinase (ADAM) proteases. NRDC has recently been revealed to be involved in the tumorigenesis of various types of cancer, including intrahepatic cholangiocarcinoma, malignant cerebral infarction, esophageal squamous cell carcinoma, and gastric cancer. However, the expression profiles and biological relevance of NRDC in pancreatic ductal adenocarcinoma have rarely been reported. Methods: We analyzed the NRDC expression profile in pancreatic ductal adenocarcinoma by enzyme-linked immunosorbent assay (ELISA) and identified NRDC as a circulating biomarker in the serum of 112 pancreatic ductal adenocarcinoma patients. The diagnostic value of NRDC was analyzed by the area under the curve (AUC) and the receiver operating characteristic (ROC) test. Results: Our results demonstrated that the clinical prognosis significance of NRDC with the clinical characteristics in pancreatic ductal adenocarcinoma (PDAC). NRDC was notably decreased in PDAC patient serum compared with the control group (p < 0.001). Furthermore, the present study found that the NRDC expression level was correlated with T grade (p < 0.001), metastasis(p < 0.001), differentiation(p < 0.001), and TNM stage (p = 0.011). Further bioinformatics analysis revealed that NRDC correlated with proliferation and migration pathways; in particular, it mediated cell-matrix adhesion-dependent activation in pancreatic ductal adenocarcinoma. Conclusions: Serum NRDC may play a useful diagnostic biomarker to evaluate the aggressive clinical features in PAAD patients.
Collapse
|
24
|
Extracellular Vesicles Carrying miR-887-3p Promote Breast Cancer Cell Drug Resistance by Targeting BTBD7 and Activating the Notch1/Hes1 Signaling Pathway. DISEASE MARKERS 2022; 2022:5762686. [PMID: 35655918 PMCID: PMC9152417 DOI: 10.1155/2022/5762686] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 12/11/2022]
Abstract
Objective Chemoresistance remains the primary reason threatening the prognosis of breast cancer (BC) patients. Extracellular vesicles (EVs) contribute to chemoresistance by carrying microRNAs (miRNAs). This study investigated the mechanism of miR-887-3p mediated by EVs in BC cell drug resistance. Methods MDA-MB-231-derived EVs were extracted and identified. BC cells were treated with different concentrations of doxorubicin, cisplatin, and fulvestrant, and the cell survival was evaluated. PKH26-labeled EVs were cocultured with BC cells, and the uptake of EVs was observed. miR-887-3p expression in BC cells and EVs was detected. After silencing miR-887-3p in MDA-MB-231 cells, BC cells were treated with EV-inhi to observe drug resistance. The target gene of miR-887-3p was predicted and verified. The levels of downstream Notch1/Hes1 pathway were detected. Xenograft experiment was conducted to evaluate the effect of EVs on the growth and drug resistance in vivo. Results MDA-MB-231-derived EVs enhanced the drug resistance of BC cells. EVs carried miR-887-3p into BC cells. miR-887-3p expression was elevated in BC cells and EVs. miR-887-3p inhibition reduced the drug resistance of BC cells. miR-887-3p targeted BTBD7. Overexpression of BTBD7 partially reversed the drug resistance of BC cells caused by EV treatment. EV treatment increased the level of Notch1/Hes1, and overexpression of BTBD7 decreased the level of Notch1/Hes1. In vivo experiments further validated the results of in vitro experiments. Conclusion EVs carrying miR-887-3p could target BTBD7 and activate the Notch1/Hes1 signaling pathway, thereby promoting BC cell drug resistance. This study may offer novel insights into BC treatment.
Collapse
|
25
|
Huang KW, Lee PC, Chao Y, Su CW, Lee IC, Lan KH, Chu CJ, Hung YP, Chen SC, Hou MC, Huang YH. Durable objective response to sorafenib and role of sequential treatment in unresectable hepatocellular carcinoma. Ther Adv Med Oncol 2022; 14:17588359221099401. [PMID: 35646162 PMCID: PMC9134461 DOI: 10.1177/17588359221099401] [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] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Background The response rate to sorafenib is limited for unresectable hepatocellular carcinoma (HCC). Little is known about the long-term outcomes of objective responders. The role of second-line therapies on the survival of sorafenib-responders is unclear. We aimed to delineate the long-term outcomes and the role of subsequent treatment after responding to sorafenib. Methods From September 2012 to December 2019, 922 patients who received sorafenib treatment for unresectable HCC were retrospectively reviewed. Of these, 21 (2.3%) achieved a complete response (CR) and 54 (5.9%) had a partial response (PR) based on mRECIST criteria. Factors associated with survivals were analyzed. Results During the median follow-up of 35.3 months, the median duration of response was 18.3 months (range: 2.3-45.5) for patients achieving CR and 10.0 months (range: 1.9-60.3) for PR. The median overall survival (OS) was 39.5 months [95% confidence interval (CI): 28.4-50.5] including values not yet estimable for CR and 25.8 months for PR. Patients who experienced treatment-related adverse events (TRAEs) had better median OS than those without (44.9 versus 18.1 months, p = 0.003). Eventually, 53 patients developed tumor progression; 30 patients received second-line systemic treatment including nivolumab (n = 8), regorafenib (n = 15), and chemotherapy (n = 7). Sorafenib-nivolumab sequential therapy provided the best median OS versus sorafenib-regorafenib and sorafenib-chemotherapy in these patients (55.8, 39.5, and 25.5 months), respectively. Conclusions The response is durable for advanced HCC patients with CR or PR to sorafenib. Subsequent immunotherapy seems to provide the best survival. This information is important for characterizing outcomes of sorafenib-responders and the choice of sequential treatment.
Collapse
Affiliation(s)
- Kuo-Wei Huang
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- Department of Medicine, Taipei City Hospital
Yang-Ming branch, Taipei
| | - Pei-Chang Lee
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - Yee Chao
- Department of Oncology, Taipei Veterans General
Hospital, Taipei
| | - Chien-Wei Su
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - I-Cheng Lee
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - Keng-Hsin Lan
- Division of Gastroenterology and Hepatology,
Department of Medicine Taipei Veterans General Hospital, Taipei
- Institute of Pharmacology, School of Medicine,
National Yang Ming Chiao Tung University, Taipei
| | - Chi-Jen Chu
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - Yi-Ping Hung
- Department of Oncology, Taipei Veterans
General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - San-Chi Chen
- Institute of Clinical Medicine, School of
Medicine, National Yang Ming Chiao Tung University, Taipei
- Department of Oncology, Taipei Veterans
General Hospital, Taipei
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- School of Medicine, National Yang Ming Chiao
Tung University, Taipei
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology,
Department of Medicine, Taipei Veterans General Hospital, Taipei
- Institute of Clinical Medicine, School of
Medicine, National Yang Ming Chiao Tung University, No.201, Sec. 2, Shipai
Road, Beitou District, 11217 Taipei
| |
Collapse
|
26
|
Xing LH, Zhuo LY, Zhang Y, Ma X, Ma ZP, Zhao YJ, Yin XP, Gao BL. DWI Combined With Hepatobiliary-Phase Enhanced Imaging Can Better Differentiate Cholangiocarcinoma From Atypical Liver Abscesses. Front Oncol 2022; 12:723089. [PMID: 35646701 PMCID: PMC9137181 DOI: 10.3389/fonc.2022.723089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To investigate the value of diffusion-weighted imaging (DWI) combined with the hepatobiliary phase (HBP) Gd-BOPTA enhancement in differentiating intrahepatic mass-forming cholangiocarcinoma (IMCC) from atypical liver abscess. Materials and Methods A retrospective analysis was performed on 43 patients with IMCCs (IMCC group) and 25 patients with atypical liver abscesses (liver abscess group). The DWI signal, the absolute value of the contrast noise ratio (│CNR│) at the HBP, and visibility were analyzed. Results A relatively high DWI signal and a relatively high peripheral signal were presented in 29 patients (67.5%) in the IMCC group, and a relatively high DWI signal was displayed in 15 patients (60.0%) in the atypical abscess group with a relatively high peripheral signal in only one (6.7%) patient and a relatively high central signal in 14 (93.3%, 14/15). A significant (P<0.001) difference existed in the pattern of signal between the two groups of patients. On T2WI, IMCC was mainly manifested by homogeneous signal (53.5%), whereas atypical liver abscesses were mainly manifested by heterogeneous signal and relatively high central signal (32%, and 64%), with a significant difference (P<0.001) in T2WI imaging presentation between the two groups. On the HBP imaging, there was a statistically significant difference in peripheral │CNR│ (P< 0.001) and visibility between two groups. The sensitivity of the HBP imaging was significantly (P=0.002) higher than that of DWI. The sensitivity and accuracy of DWI combined with enhanced HBP imaging were significantly (P=0.002 and P<0.001) higher than those of either HBP imaging or DWI alone. Conclusion Intrahepatic mass-forming cholangiocarcinoma and atypical liver abscesses exhibit different imaging signals, and combination of DWI and hepatobiliary-phase enhanced imaging has higher sensitivity and accuracy than either technique in differentiating intrahepatic mass-forming cholangiocarcinoma from atypical liver abscesses.
Collapse
Affiliation(s)
- Li-Hong Xing
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
- School of Clinical Medicine of Hebei University, Baoding, China
| | - Li-Yong Zhuo
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| | - Yu Zhang
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| | - Xi Ma
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| | - Ze-Peng Ma
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| | - Ying-Jia Zhao
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| | - Xiao-Ping Yin
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
- *Correspondence: Xiao-Ping Yin, ; orcid.org/0000-0002-7386-1069
| | - Bu-Lang Gao
- Department of CT/MRI Room, Affiliated Hospital of Hebei University, and Key Laboratory of Cancer Radiotherapy and Chemotherapy Mechanism and Regulations, Baoding, China
| |
Collapse
|
27
|
Frisone D, Friedlaender A, Addeo A, Tsantoulis P. The Landscape of Immunotherapy Resistance in NSCLC. Front Oncol 2022; 12:817548. [PMID: 35515125 PMCID: PMC9066487 DOI: 10.3389/fonc.2022.817548] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Lung cancer is the leading cause of cancer mortality worldwide. Immunotherapy has demonstrated clinically significant benefit for non-small-cell lung cancer, but innate (primary) or acquired resistance remains a challenge. Criteria for a uniform clinical definition of acquired resistance have been recently proposed in order to harmonize the design of future clinical trials. Several mechanisms of resistance are now well-described, including the lack of tumor antigens, defective antigen presentation, modulation of critical cellular pathways, epigenetic changes, and changes in the tumor microenvironment. Host-related factors, such as the microbiome and the state of immunity, have also been examined. New compounds and treatment strategies are being developed to target these mechanisms with the goal of maximizing the benefit derived from immunotherapy. Here we review the definitions of resistance to immunotherapy, examine its underlying mechanisms and potential corresponding treatment strategies. We focus on recently published clinical trials and trials that are expected to deliver results soon. Finally, we gather insights from recent preclinical discoveries that may translate to clinical application in the future.
Collapse
Affiliation(s)
- Daniele Frisone
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Alex Friedlaender
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland.,Department of Oncology, Clinique Generale Beaulieu, Geneva, Switzerland
| | - Alfredo Addeo
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland.,Department of Oncology, Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Petros Tsantoulis
- Department of Oncology, Geneva University Hospital, Geneva, Switzerland.,Department of Oncology, Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
28
|
D'Alessandro R, Refolo MG, Schirizzi A, De Leonardis G, Donghia R, Guerra V, Giannelli G, Lolli IR, Laterza MM, De Vita F, Messa C, Lotesoriere C. Variations in Circulating Levels of Angiopoietin-2 Over Time Are Predictive of Ramucirumab-Paclitaxel Therapy Outcome in Advanced Gastric Cancer: Results of Prospective Study. Front Oncol 2022; 12:862116. [PMID: 35463372 PMCID: PMC9019360 DOI: 10.3389/fonc.2022.862116] [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: 01/25/2022] [Accepted: 02/25/2022] [Indexed: 11/13/2022] Open
Abstract
The combination of paclitaxel and ramucirumab is the second-line therapy of choice in the treatment of advanced gastric cancer. To date, no biomarkers are available in gastric cancer to predict the outcome of antiangiogenic therapy. The present prospective study included 35 patients undergoing second-line therapy with ramucirumab and paclitaxel. Serum samples were systematically collected from the beginning of therapy and at each cycle until disease progression. Multiplex analysis of a panel of angiogenic factors identified markers for which the changes at defined time intervals were significantly different in patients with progression-free survival ≤3 (Rapid Progression Group) compared to those with progression-free survival >3 (Control Disease Group). Comparative analysis revealed significantly different results in the two groups of patients for VEGFC and Angiopoietin-2, both involved in angiogenesis and lymphangiogenesis. VEGFC increased in the progressive-disease group, while it decreased in the control-disease group. This decrease persisted beyond the third cycle, and it was statistically significant compared to the basal level in patients with longer progression-free survival. Angiopoietin-2 decreased significantly after 2 months of therapy. At progression time, there was a significant increase in VEGFC and Angiopoietin-2, suggesting the activation pathways counteracting the blockade of VEGFR2 by ramucirumab. Overall results showed that a greater change in VEGFC and Angiopoietin-2 levels measured at the beginning of the third cycle of therapy corresponded to a lower risk of progression and thus to longer progression-free survival.
Collapse
Affiliation(s)
- Rosalba D'Alessandro
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Maria Grazia Refolo
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Annalisa Schirizzi
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Giampiero De Leonardis
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Rossella Donghia
- National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Vito Guerra
- National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology "Saverio de Bellis", Research Hospital, Castellana Grotte, Italy
| | - Ivan Roberto Lolli
- Medical Oncology Unit, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Maria Maddalena Laterza
- Complex Operating Unit Oncologia, Local Health Authority Napoli 2 Nord, P.O. "S.M. delle Grazie", Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Precision Medicine, School of Medicine, University of Study of Campania "Luigi Vanvitelli", Naples, Italy
| | - Caterina Messa
- Laboratory of Experimental Oncology, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| | - Claudio Lotesoriere
- Medical Oncology Unit, National Institute of Gastroenterology, "Saverio de Bellis" Research Hospital, Castellana Grotte, Italy
| |
Collapse
|
29
|
Endothelial Cell Metabolism in Vascular Functions. Cancers (Basel) 2022; 14:cancers14081929. [PMID: 35454836 PMCID: PMC9031281 DOI: 10.3390/cancers14081929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Recent findings in the field of vascular biology are nourishing the idea that targeting the endothelial cell metabolism may be an alternative strategy to antiangiogenic therapy, as well as a novel therapeutic approach for cardiovascular disease. Deepening the molecular mechanisms regulating how ECs re-adapt their metabolic status in response to the changeable conditions of the tissue microenvironment may be beneficial to develop novel innovative treatments to counteract the aberrant growth of vasculature. Abstract The endothelium is the innermost layer of all blood and lymphatic vessels composed of a monolayer of specialized endothelial cells (ECs). It is regarded as a dynamic and multifunctional endocrine organ that takes part in essential processes, such as the control of blood fluidity, the modulation of vascular tone, the regulation of immune response and leukocyte trafficking into perivascular tissues, and angiogenesis. The inability of ECs to perform their normal biological functions, known as endothelial dysfunction, is multi-factorial; for instance, it implicates the failure of ECs to support the normal antithrombotic and anti-inflammatory status, resulting in the onset of unfavorable cardiovascular conditions such as atherosclerosis, coronary artery disease, hypertension, heart problems, and other vascular pathologies. Notably, it is emerging that the ability of ECs to adapt their metabolic status to persistent changes of the tissue microenvironment could be vital for the maintenance of vascular functions and to prevent adverse vascular events. The main purpose of the present article is to shed light on the unique metabolic plasticity of ECs as a prospective therapeutic target; this may lead to the development of novel strategies for cardiovascular diseases and cancer.
Collapse
|
30
|
Germline Variants in Angiogenesis-Related Genes Contribute to Clinical Outcome in Head and Neck Squamous Cell Carcinoma. Cancers (Basel) 2022; 14:cancers14071844. [PMID: 35406617 PMCID: PMC8997703 DOI: 10.3390/cancers14071844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/28/2022] [Accepted: 04/01/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary A high risk of relapse and treatment resistance are among the major challenges in locally advanced head and neck squamous cell carcinoma (HNSCC). Data show that common germline alterations in genes regulating angiogenesis may modulate treatment sensitivity, cancer progression, and prognosis, but relatively little is known about their role in HNSCC. Thus, our goal was to examine the effect of variation in these genes on survival outcomes in HNSCC patients receiving radiotherapy and cisplatin-based chemoradiotherapy. We identified genetic variants significantly affecting therapy results, constituting independent prognostic factors in these patients. Our results suggest that some polymorphisms in angiogenesis genes may be determinants of treatment efficacy and tumor aggressiveness in HNSCC, which may be of importance in standard therapy. These findings emphasize the potential value of the host genetic profile related to angiogenesis in assessing the risk of treatment failure. Abstract Fibroblast growth factor (FGF)/FGF receptor (FGFR), and platelet-derived growth factor (PDGF)/PDGF receptor (PDGFR) systems, as well as some matrix metalloproteinases (MMPs) and their tissue inhibitors (TIMPs), are involved in various steps of angiogenesis. Data indicate that common germline variations in angiogenesis-regulating genes may modulate therapy results and cancer progression. However, whether these variants affect clinical outcome in head and neck squamous cell carcinoma (HNSCC) is unclear. Hence, we assessed the relationship between FGF/FGFR, PDGF/PDGFR, MMP, and TIMP genetic variants and treatment outcomes in HNSCC patients receiving radiotherapy (RT) alone or combined with cisplatin-based chemotherapy. In multivariate analysis, FGF2 rs1048201 CC homozygotes showed a higher risk of death (p = 0.039), while PDGFRA rs2228230 T was strongly associated with an increased risk of locoregional relapse (HR 2.49, p = 0.001) in the combination treatment subgroup. In the RT alone subset, MMP2 rs243865 TT carriers had a higher risk of locoregional recurrence (HR 2.92, p = 0.019), whereas PDGFRB rs246395 CC homozygotes were at increased risk of metastasis (HR 3.06, p = 0.041). The MMP2 rs7201 C and TIMP2 rs7501477 T were associated with a risk of locoregional failure in the entire cohort (p = 0.032 and 0.045, respectively). Furthermore, rs1048201, rs2228230, rs246395, rs243865, rs7201, and rs7201/rs7501477 were independent indicators of an unfavorable outcome. This study demonstrates that the FGF2, PDGFRA, PDGFRB, MMP2, and TIMP2 variants may contribute to treatment failure and poor prognosis in HNSCC.
Collapse
|
31
|
Timmins MA, Ringshausen I. Transforming Growth Factor-Beta Orchestrates Tumour and Bystander Cells in B-Cell Non-Hodgkin Lymphoma. Cancers (Basel) 2022; 14:1772. [PMID: 35406544 PMCID: PMC8996985 DOI: 10.3390/cancers14071772] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/23/2022] [Indexed: 12/15/2022] Open
Abstract
Transforming growth factor-beta (TGFB) is a critical regulator of normal haematopoiesis. Dysregulation of the TGFB pathway is associated with numerous haematological malignancies including myelofibrosis, acute myeloid leukaemia, and lymphoid disorders. TGFB has classically been seen as a negative regulator of proliferation in haematopoiesis whilst stimulating differentiation and apoptosis, as required to maintain homeostasis. Tumours frequently develop intrinsic resistant mechanisms to homeostatic TGFB signalling to antagonise its tumour-suppressive functions. Furthermore, elevated levels of TGFB enhance pathogenesis through modulation of the immune system and tumour microenvironment. Here, we review recent advances in the understanding of TGFB signalling in B-cell malignancies with a focus on the tumour microenvironment. Malignant B-cells harbour subtype-specific alterations in TGFB signalling elements including downregulation of surface receptors, modulation of SMAD signalling proteins, as well as genetic and epigenetic aberrations. Microenvironmental TGFB generates a protumoural niche reprogramming stromal, natural killer (NK), and T-cells. Increasingly, evidence points to complex bi-directional cross-talk between cells of the microenvironment and malignant B-cells. A greater understanding of intercellular communication and the context-specific nature of TGFB signalling may provide further insight into disease pathogenesis and future therapeutic strategies.
Collapse
Affiliation(s)
- Matthew A. Timmins
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| | - Ingo Ringshausen
- Wellcome Trust/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AH, UK;
- Department of Haematology, Addenbrooke’s Hospital, Cambridge University Hospital, Cambridge CB2 0AH, UK
| |
Collapse
|
32
|
Azzam SK, Alsafar H, Sajini AA. FTO m6A Demethylase in Obesity and Cancer: Implications and Underlying Molecular Mechanisms. Int J Mol Sci 2022; 23:ijms23073800. [PMID: 35409166 PMCID: PMC8998816 DOI: 10.3390/ijms23073800] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 12/20/2022] Open
Abstract
Fat mass and obesity-associated protein (FTO) is the first reported RNA N6-methyladenosine (m6A) demethylase in eukaryotic cells. m6A is considered as the most abundant mRNA internal modification, which modulates several cellular processes including alternative splicing, stability, and expression. Genome-wide association studies (GWAS) identified single-nucleotide polymorphisms (SNPs) within FTO to be associated with obesity, as well as cancer including endometrial cancer, breast cancer, pancreatic cancer, and melanoma. Since the initial classification of FTO as an m6A demethylase, various studies started to unravel a connection between FTO’s demethylase activity and the susceptibility to obesity on the molecular level. FTO was found to facilitate adipogenesis, by regulating adipogenic pathways and inducing pre-adipocyte differentiation. FTO has also been investigated in tumorigenesis, where emerging studies suggest m6A and FTO levels are dysregulated in various cancers, including acute myeloid leukemia (AML), glioblastoma, cervical squamous cell carcinoma (CSCC), breast cancer, and melanoma. Here we review the molecular bases of m6A in tumorigenesis and adipogenesis while highlighting the controversial role of FTO in obesity. We provide recent findings confirming FTO’s causative link to obesity and discuss novel approaches using RNA demethylase inhibitors as targeted oncotherapies. Our review aims to confirm m6A demethylation as a risk factor in obesity and provoke new research in FTO and human disorders.
Collapse
Affiliation(s)
- Sarah Kassem Azzam
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Healthcare Engineering Innovation Center (HEIC), Department of Biomedical Engineering, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Habiba Alsafar
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Genetics and Molecular Biology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Emirates Bio-Research Center, Ministry of Interior, Abu Dhabi P.O. Box 389, United Arab Emirates
| | - Abdulrahim A. Sajini
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi P.O. Box 127788, United Arab Emirates; (S.K.A.); (H.A.)
- Healthcare Engineering Innovation Center (HEIC), Department of Biomedical Engineering, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Correspondence:
| |
Collapse
|
33
|
Shin HJ, Gil M, Lee IS. Association of Elevated Expression Levels of COL4A1 in Stromal Cells with an Immunosuppressive Tumor Microenvironment in Low-Grade Glioma, Pancreatic Adenocarcinoma, Skin Cutaneous Melanoma, and Stomach Adenocarcinoma. J Pers Med 2022; 12:534. [PMID: 35455650 PMCID: PMC9029283 DOI: 10.3390/jpm12040534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Aberrant expression of collagen type IV alpha chain 1 (COL4A1) can influence tumor cell behavior. To examine the association of COL4A1 expression in the tumor microenvironment (TME) with tumor progression, we performed bioinformatics analyses of The Cancer Genome Atlas RNA sequencing and RNA microarray datasets available in public databases and identified upregulated COL4A1 expression in most examined tumor types compared to their normal counterparts. The elevated expression of COL4A1 was correlated with low survival rates of patients with low-grade glioma, pancreatic adenocarcinoma, skin cutaneous melanoma, and stomach adenocarcinoma, thus suggesting its potential use as a biomarker for the poor prognosis of these tumors. However, COL4A1 was mostly expressed in adjacent stromal cells, such as cancer-associated fibroblasts (CAFs) and endothelial cells. Additionally, COL4A1 expression was highly correlated with the signatures of CAFs and endothelial cells in all four tumor types. The expression of marker genes for the infiltration of pro-tumoral immune cells, such as Treg, M2, and TAM, and those of immunosuppressive cytokines exhibited very strong positive correlations with COL4A1 expression. Collectively, our data suggest that COL4A1 overexpression in stromal cells may be a potential regulator of tumor-supporting TME composition associated with poor prognosis.
Collapse
Affiliation(s)
- Hyo-Jae Shin
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea;
| | - Minchan Gil
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea
| | - Im-Soon Lee
- Department of Biological Sciences, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea;
| |
Collapse
|
34
|
Sakano Y, Noda T, Kobayashi S, Sasaki K, Iwagami Y, Yamada D, Tomimaru Y, Akita H, Gotoh K, Takahashi H, Asaoka T, Tanemura M, Wada H, Doki Y, Eguchi H. Tumor endothelial cell-induced CD8(+) T-cell exhaustion via GPNMB in hepatocellular carcinoma. Cancer Sci 2022; 113:1625-1638. [PMID: 35289033 PMCID: PMC9128167 DOI: 10.1111/cas.15331] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 11/26/2022] Open
Abstract
Tumor endothelial cells (TECs) promote tumor angiogenesis and regulate cytotoxic T cells in the tumor microenvironment. However, the roles of TECs for tumor‐infiltrating T‐cell in hepatocellular carcinoma (HCC) is still unknown. Here, we aimed to investigate how TECs influenced tumor growth and immune responses of HCC focusing on CD8+ T‐cell infiltration and exhaustion. First, TECs were isolated from subcutaneous HCC tumors with murine HCC cell lines (BNL‐T) with magnetic selection of CD31+ cells, and normal endothelial cells (NECs) were isolated from normal liver. Second, immunocompetent mice were injected with BNL‐T alone, BNL‐T + NECs, or BNL‐T + TECs for tumor formation, and the functions and exhaustion of tumor‐infiltrating CD8+ T cells were evaluated. The mice injected with BNL‐T + TEC showed rapid tumorigenesis and a decrease in the number of infiltrating CD8+ T cells. In addition, the percentage of CD8+ T‐cell exhaustion was significantly higher in tumors from the administration of BNL‐T + TEC. Third, the next‐generation sequencing on TECs was performed to identify mRNAs that might be a novel treatment target. The molecule of glycoprotein nonmetastatic melanoma protein B (GPNMB) was identified and the functions of GPNMB was analyzed by silencing of GPNMB expression using small interfering RNAs. The silencing of GPNMB expression in TECs induced the suppression of tumor growth and T‐cell exhaustion. In conclusion, TECs induced tumor‐infiltrating T‐cell exhaustion via GPNMB expression and GPNMB might be a novel therapeutic target in HCC.
Collapse
Affiliation(s)
- Yoshihiro Sakano
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takehiro Noda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Shogo Kobayashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kazuki Sasaki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshifumi Iwagami
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Daisaku Yamada
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yoshito Tomimaru
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Akita
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Kunihito Gotoh
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidenori Takahashi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tadafumi Asaoka
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Surgery, Osaka Police Hospital, Osaka, Japan
| | - Masahiro Tanemura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Surgery, Rinku General Medical Center, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University
| | - Yuichiro Doki
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidetoshi Eguchi
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
35
|
Wang X, Xu W, Hu X, Yang X, Zhang M. The Prognostic Role of Glycemia in Patients With Pancreatic Carcinoma: A Systematic Review and Meta-Analysis. Front Oncol 2022; 12:780909. [PMID: 35223469 PMCID: PMC8866248 DOI: 10.3389/fonc.2022.780909] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Background Fasting blood glucose and glycated hemoglobin (HbA1c) levels are associated with the risk of pancreatic cancer. Aim To examine the relationship between perioperative glucose and HbA1c levels and prognosis in patients with pancreatic cancer. Methods PubMed, Embase, and the Cochrane Library were queried for potentially eligible studies published up to May 2021. The exposures were perioperative fasting glucose and HbA1c levels. The primary outcome was survival. The secondary outcome was complications. All analyses were performed using the random-effects model. Results Ten studies (48,424 patients) were included. The pre-operative (HR=1.10, 95%CI: 0.89-1.35; I2 = 45.1%, Pheterogeneity=0.078) and postoperative (HR=1.19, 95%CI: 0.92-1.54; I2 = 67.9%, Pheterogeneity=0.001) blood glucose levels were not associated with the survival to pancreatic cancer. Similar results were observed for HbA1c (HR=1.09, 95%CI: 0.75-1.58; I2 = 64.2%, Pheterogeneity=0.039), fasting blood glucose (FBG)/HbA1c (HR=1.16, 95%CI: 0.67-1.68; I2 = 0.0%, Pheterogeneity=0.928), and FBG (HR=1.75, 95%CI: 0.81-3.75; I2 = 79.4%, Pheterogeneity=0.008). Pre-operative blood glucose levels were not associated with postoperative complications (OR=0.90, 95%CI: 0.52-1.56), but postoperative glucose levels were associated with postoperative complications (OR=3.06, 95%CI: 1.88-4.97; I2 = 0.0%, Pheterogeneity=0.619). Conclusion Blood glucose, FBG, and HbA1c levels are not associated with the survival of patients with pancreatic cancer. Postoperative blood glucose levels could predict postoperative complications.
Collapse
Affiliation(s)
- Xiaofang Wang
- Department of Clinical Oncology and Department of Hospice Care, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wanfeng Xu
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaoru Hu
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xianghong Yang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingming Zhang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
36
|
Solimando AG, Kalogirou C, Krebs M. Angiogenesis as Therapeutic Target in Metastatic Prostate Cancer - Narrowing the Gap Between Bench and Bedside. Front Immunol 2022; 13:842038. [PMID: 35222436 PMCID: PMC8866833 DOI: 10.3389/fimmu.2022.842038] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/21/2022] [Indexed: 12/14/2022] Open
Abstract
Angiogenesis in metastatic castration-resistant prostate cancer (mCRPC) has been extensively investigated as a promising druggable biological process. Nonetheless, targeting angiogenesis has failed to impact overall survival (OS) in patients with mCRPC despite promising preclinical and early clinical data. This discrepancy prompted a literature review highlighting the tumor heterogeneity and biological context of Prostate Cancer (PCa). Narrowing the gap between the bench and bedside appears critical for developing novel therapeutic strategies. Searching clinicaltrials.gov for studies examining angiogenesis inhibition in patients with PCa resulted in n=20 trials with specific angiogenesis inhibitors currently recruiting (as of September 2021). Moreover, several other compounds with known anti-angiogenic properties - such as Metformin or Curcumin - are currently investigated. In general, angiogenesis-targeting strategies in PCa include biomarker-guided treatment stratification - as well as combinatorial approaches. Beyond established angiogenesis inhibitors, PCa therapies aiming at PSMA (Prostate Specific Membrane Antigen) hold the promise to have a substantial anti-angiogenic effect - due to PSMA´s abundant expression in tumor vasculature.
Collapse
Affiliation(s)
- Antonio Giovanni Solimando
- Department of Biomedical Sciences and Human Oncology, Section of Internal Medicine "G. Baccelli", University of Bari Medical School, Bari, Italy.,Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Charis Kalogirou
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany
| | - Markus Krebs
- Department of Urology and Pediatric Urology, University Hospital Würzburg, Würzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
37
|
Identifying Potential New Gene Expression-Based Biomarkers in the Peripheral Blood Mononuclear Cells of Hepatitis B-Related Hepatocellular Carcinoma. Can J Gastroenterol Hepatol 2022; 2022:9541600. [PMID: 35265561 PMCID: PMC8901362 DOI: 10.1155/2022/9541600] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/13/2021] [Accepted: 01/22/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE The analysis of the gene expression of peripheral blood mononuclear cells (PBMCs) is important to clarify the pathogenesis of hepatocellular carcinoma (HCC) and the detection of suitable biomarkers. The purpose of this investigation was to use RNA-sequencing to screen the appropriate differentially expressed genes (DEGs) in the PBMCs for the HCC. METHODS The comprehensive transcriptome of extracted RNA of PBMC (n = 20) from patients with chronic hepatitis B (CHB), liver cirrhosis, and early stage of HCC (5 samples per group) was carried out using RNA-sequencing. All raw RNA-sequencing data analyses were performed using conventional RNA-sequencing analysis tools. Next, gene ontology (GO) analyses were carried out to elucidate the biological processes of DEGs. Finally, relative transcript abundance of selected DEGs was verified using qRT-PCR on additional validation groups. RESULTS Specifically, 13, 1262, and 1450 DEGs were identified for CHB, liver cirrhosis, and HCC, when compared with the healthy controls. GO enrichment analysis indicated that HCC is closely related to the immune response. Seven DEGs (TYMP, TYROBP, CD14, TGFBI, LILRA2, GNLY, and GZMB) were common to HCC, cirrhosis, and CHB when compared to healthy controls. The data revealed that the expressions of these 7 DEGs were consistent with those from the RNA-sequencing results. Also, the expressions of 7 representative genes that had higher sensitivity were obtained by receiver operating characteristic analysis, which indicated their important diagnostic accuracy for HBV-HCC. CONCLUSION This study provides us with new horizons into the biological process and potential prospective clinical diagnosis and prognosis of HCC in the near future.
Collapse
|
38
|
Forouzannezhad P, Maes D, Hippe DS, Thammasorn P, Iranzad R, Han J, Duan C, Liu X, Wang S, Chaovalitwongse WA, Zeng J, Bowen SR. Multitask Learning Radiomics on Longitudinal Imaging to Predict Survival Outcomes following Risk-Adaptive Chemoradiation for Non-Small Cell Lung Cancer. Cancers (Basel) 2022; 14:1228. [PMID: 35267535 PMCID: PMC8909466 DOI: 10.3390/cancers14051228] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 11/16/2022] Open
Abstract
Medical imaging provides quantitative and spatial information to evaluate treatment response in the management of patients with non-small cell lung cancer (NSCLC). High throughput extraction of radiomic features on these images can potentially phenotype tumors non-invasively and support risk stratification based on survival outcome prediction. The prognostic value of radiomics from different imaging modalities and time points prior to and during chemoradiation therapy of NSCLC, relative to conventional imaging biomarker or delta radiomics models, remains uncharacterized. We investigated the utility of multitask learning of multi-time point radiomic features, as opposed to single-task learning, for improving survival outcome prediction relative to conventional clinical imaging feature model benchmarks. Survival outcomes were prospectively collected for 45 patients with unresectable NSCLC enrolled on the FLARE-RT phase II trial of risk-adaptive chemoradiation and optional consolidation PD-L1 checkpoint blockade (NCT02773238). FDG-PET, CT, and perfusion SPECT imaging pretreatment and week 3 mid-treatment was performed and 110 IBSI-compliant pyradiomics shape-/intensity-/texture-based features from the metabolic tumor volume were extracted. Outcome modeling consisted of a fused Laplacian sparse group LASSO with component-wise gradient boosting survival regression in a multitask learning framework. Testing performance under stratified 10-fold cross-validation was evaluated for multitask learning radiomics of different imaging modalities and time points. Multitask learning models were benchmarked against conventional clinical imaging and delta radiomics models and evaluated with the concordance index (c-index) and index of prediction accuracy (IPA). FDG-PET radiomics had higher prognostic value for overall survival in test folds (c-index 0.71 [0.67, 0.75]) than CT radiomics (c-index 0.64 [0.60, 0.71]) or perfusion SPECT radiomics (c-index 0.60 [0.57, 0.63]). Multitask learning of pre-/mid-treatment FDG-PET radiomics (c-index 0.71 [0.67, 0.75]) outperformed benchmark clinical imaging (c-index 0.65 [0.59, 0.71]) and FDG-PET delta radiomics (c-index 0.52 [0.48, 0.58]) models. Similarly, the IPA for multitask learning FDG-PET radiomics (30%) was higher than clinical imaging (26%) and delta radiomics (15%) models. Radiomics models performed consistently under different voxel resampling conditions. Multitask learning radiomics for outcome modeling provides a clinical decision support platform that leverages longitudinal imaging information. This framework can reveal the relative importance of different imaging modalities and time points when designing risk-adaptive cancer treatment strategies.
Collapse
Affiliation(s)
- Parisa Forouzannezhad
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Dominic Maes
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
| | - Phawis Thammasorn
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Reza Iranzad
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jie Han
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - Chunyan Duan
- Department of Mechanical Engineering, Tongji University, Shanghai 200092, China;
| | - Xiao Liu
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Shouyi Wang
- Department of Industrial, Manufacturing, and System Engineering, University of Texas, Arlington, TX 76019, USA; (J.H.); (S.W.)
| | - W. Art Chaovalitwongse
- Department of Industrial Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (P.T.); (R.I.); (X.L.); (W.A.C.)
| | - Jing Zeng
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
| | - Stephen R. Bowen
- Department of Radiation Oncology, School of Medicine, University of Washington, Seattle, WA 98195, USA; (P.F.); (D.M.); (J.Z.)
- Department of Radiology, School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
39
|
GGT5 Is an Independent Prognostic Biomarker in Stomach Adenocarcinoma. Can J Gastroenterol Hepatol 2022; 2022:9983351. [PMID: 35257007 PMCID: PMC8898138 DOI: 10.1155/2022/9983351] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer is one of the cancers with the highest incidence in the world. Gamma-glutamyltransferase 5 (GGT5) is expressed in different cancers and its role in cancers remains unclear. In this study, we aimed to evaluate the value of GGT5 in stomach adenocarcinoma (STAD). In TCGA, patients with high GGT5 expression had poor overall survival (P=0.006). Based on GSE62254, high expression of GGT5 was associated with poor OS (P=0.014) and PFS (P=0.042). The same result was observed in GSE14210. We further discovered that GGT5 expression was associated with stage, grade, and T stage. Further prognostic analysis of GGT5 showed that GGT5 was associated with prognosis in both univariate analysis (P=0.032) and multivariate analysis (P=0.029). We used gene set enrichment analysis (GSEA) to explore the possible mechanism of GGT5. GSEA suggests that overexpression of GGT5 may be involved in leukocyte transendothelial migration, JAK-STAT signaling pathway, MAPK signaling pathway, and melanoma. The high-expression GGT5 group had higher concentrations of M2 macrophages, T cell regulators, and monocytes, but the contents of plasma cells and M1 macrophages were higher in the low-expression GGT5 group. The results showed that the ESTIMATEScore, ImmuneScore, and StromalScore of the high-expression GGT5 group were higher than those of the low-expression GGT5 group. PD1 and CTLA4 expression levels were higher in the high-expression GGT5 group. The high-expression GGT5 group may be more effective for immunotherapy. These results suggested that GGT5 could be a potential prognostic molecular predictor in STAD.
Collapse
|
40
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
41
|
Pan Y, Si H, Deng G, Chen S, Zhang N, Zhou Q, Wang Z, Dai G. A Composite Biomarker of Derived Neutrophil–Lymphocyte Ratio and Platelet–Lymphocyte Ratio Correlates With Outcomes in Advanced Gastric Cancer Patients Treated With Anti-PD-1 Antibodies. Front Oncol 2022; 11:798415. [PMID: 35251952 PMCID: PMC8895371 DOI: 10.3389/fonc.2021.798415] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/29/2021] [Indexed: 12/25/2022] Open
Abstract
BackgroundThe highly heterogeneous characteristics of GC may limit the accuracy of a single biomarker for screening populations benefiting from immunotherapy. However, the combination of multiple indicators can provide more directed information for the detection of potential immune benefit subgroups. At present, there are no recognized complex indexes to identify advanced GC (AGC) in patients who likely benefited from immunotherapy. The objective of this research is to explore whether the composite biomarker of derived neutrophil–lymphocyte ratio (dNLR) and platelet–lymphocyte ratio (PLR) can be used as a reliable prognostic factor for the survival of AGC patients receiving immunotherapy.MethodsFrom December 2014 to May 2021, a total 238 AGC patients at a single Center were included in this retrospective cohort research study. The cutoff value of dNLR was obtained by the ROC curves to predict the disease progression rate at the 8th month and the cutoff value of PLR was estimated by the median value. The cutoff values of dNLR and PLR were 1.95 and 163.63, respectively. The high levels of dNLR (≥1.95) and PLR (≥163.63) were considered to be risk factors. Based on these two risk factors, patients were categorized into 3 groups: the risk factor number for the “good” group was 0, that for the “intermediate” group was 1, and that for the “poor” group was 2. The subjects were divided into two groups: dNLR/PLR-good and dNLR/PLR-intermediate/poor.ResultsOf the 238 patients, the median overall survival (mOS) and progression-free survival (mPFS) were 12.5 and 4.7 months, respectively. Multivariate analysis revealed that the good dNLR/PLR group was independently associated with better prognosis. The intermediate/poor dNLR/PLR group was independently correlated with an over 1.4 times greater risk of disease progression (4.1 months vs. 5.5 months; p = 0.016) and an over 1.54 times greater risk of death (11.1 months vs. 26.3 months; p = 0.033) than the good dNLR/PLR group. However, no clear differences in the disease control rate (DCR) and overall response rate (ORR) were observed between the intermediate/poor dNLR/PLR group and the good dNLR/PLR group (51.5% vs. 56.3%, 26.3% vs. 29.6%; p = 0.494, p = 0.609).ConclusionOur study firstly verifies that the composite biomarker of dNLR and PLR is an independent prognostic factor affecting survival of advanced AGC patients receiving immunotherapy. It may be difficult for patients with the intermediate/poor dNLR/PLR group to benefit from immunotherapy.
Collapse
Affiliation(s)
- Yuting Pan
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Haiyan Si
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Guochao Deng
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Shiyun Chen
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Nan Zhang
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Qian Zhou
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - ZhiKuan Wang
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: ZhiKuan Wang, ; Guanghai Dai,
| | - Guanghai Dai
- Chinese People’s Liberation Army Medical School, Beijing, China
- Medical Oncology Department, The First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- *Correspondence: ZhiKuan Wang, ; Guanghai Dai,
| |
Collapse
|
42
|
Benitez-Amaro A, Martínez-Bosch N, Manero-Rupérez N, Claudi L, La Chica Lhoëst MT, Soler M, Ros-Blanco L, Navarro P, Llorente-Cortés V. Peptides against Low Density Lipoprotein (LDL) Aggregation Inhibit Intracellular Cholesteryl Ester Loading and Proliferation of Pancreatic Tumor Cells. Cancers (Basel) 2022; 14:cancers14040890. [PMID: 35205638 PMCID: PMC8869901 DOI: 10.3390/cancers14040890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Dyslipidemia is a modifiable risk factor for pancreatic ductal adenocarcinoma (PDAC), one of the most lethal cancers. A key component of dyslipidemia is a high level of small and dense low-density lipoproteins (LDLs). These LDLs have a high probability to be entrapped and modified (aggregated) in the extracellular matrix (ECM), becoming a source of cholesterol for tumor cells. However, the effect of aggregated LDLs on tumor progression has been unexplored. The aim of this work was to determine the effect of modified LDLs on intracellular cholesteryl ester/free cholesterol ratio (CE/FC) and cancer cell growth, and the efficacy of peptides designed to inhibit LDL aggregation on these processes. Our results show that aggregated LDL upregulates the intracellular CE/FC ratio and cell growth in pancreatic cancer and that these upregulatory effects were blocked by peptides against LDL aggregation. We propose that anti-LDL aggregation peptides deserve to be further investigated as anti-tumoral strategies. Abstract Dyslipidemia, metabolic disorders and/or obesity are postulated as risk factors for pancreatic ductal adenocarcinoma (PDAC). The majority of patients with these metabolic alterations have low density lipoproteins (LDLs) with increased susceptibility to become aggregated in the extracellular matrix (ECM). LDL aggregation can be efficiently inhibited by low-density lipoprotein receptor-related protein 1 (LRP1)-based peptides. The objectives of this work were: (i) to determine if aggregated LDLs affect the intracellular cholesteryl ester (CE)/free cholesterol (FC) ratio and/or the tumor pancreatic cell proliferation, using sphingomyelinase-modified LDL particles (Aggregated LDL, AgLDL); and (ii) to test whether LRP1-based peptides, highly efficient against LDL aggregation, can interfere in these processes. For this, we exposed human pancreatic cancer cell lines (PANC-1, RWP-1 and Capan-1) to native (nLDL) or AgLDLs in the absence or presence of LRP1-based peptides (DP3) or irrelevant peptides (IP321). Results of thin-layer chromatography (TLC) following lipid extraction indicate that AgLDLs induce a higher intracellular CE/FC ratio than nLDL, and that DP3 but not IP321 counteracts this effect. AgLDLs also increase PANC-1 cell proliferation, which is inhibited by the DP3 peptide. Our results indicate that AgLDL-induced intracellular CE accumulation plays a crucial role in the proliferation of pancreatic tumor cell lines. Peptides with anti-LDL aggregation properties may thus exhibit anti-tumor effects.
Collapse
Affiliation(s)
- Aleyda Benitez-Amaro
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.-A.); (L.C.); (M.T.L.C.L.)
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
| | - Neus Martínez-Bosch
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain; (N.M.-B.); (N.M.-R.)
| | - Noemí Manero-Rupérez
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain; (N.M.-B.); (N.M.-R.)
| | - Lene Claudi
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.-A.); (L.C.); (M.T.L.C.L.)
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
| | - Maria Teresa La Chica Lhoëst
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.-A.); (L.C.); (M.T.L.C.L.)
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
| | - Marta Soler
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
| | - Lia Ros-Blanco
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
| | - Pilar Navarro
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.-A.); (L.C.); (M.T.L.C.L.)
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), Unidad Asociada IIBB-CSIC, 08003 Barcelona, Spain; (N.M.-B.); (N.M.-R.)
- August Pi Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain
- Correspondence: (P.N.); (V.L.-C.)
| | - Vicenta Llorente-Cortés
- Institute of Biomedical Research of Barcelona (IIBB), Spanish National Research Council (CSIC), 08036 Barcelona, Spain; (A.B.-A.); (L.C.); (M.T.L.C.L.)
- Biomedical Research Institute Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain; (M.S.); (L.R.-B.)
- CIBERCV, Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (P.N.); (V.L.-C.)
| |
Collapse
|
43
|
Wu L, Zhao X, Ma H, Zhang L, Li X. Discoidin Domain Receptor 1, a Potential Biomarker and Therapeutic Target in Hepatocellular Carcinoma. Int J Gen Med 2022; 15:2037-2044. [PMID: 35237068 PMCID: PMC8882470 DOI: 10.2147/ijgm.s348110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 02/10/2022] [Indexed: 11/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is still one of the most lethal human cancers in the world due to its high degree of malignancy, easy invasion and metastasis, poor therapeutic effect and poor prognosis. Nowadays, there is no very effective diagnosis and treatment method. It is crucial to elucidate the underlying pathogenesis and mechanisms of HCC for developing new and effective diagnostic/prognostic biomarkers and therapies. Discoidin domain receptors (DDRs) belong to the family of transmembrane receptor tyrosine kinases (RTKs) and are recognized as playing central regulatory roles in a variety of high incidence human diseases, including tumors. DDRs have two members, DDR1 and DDR2. The role of DDR1 in several tumors has been extensively studied, and many researchers have identified it as a powerful candidate target for the development of functional and effective tumor treatment inhibitors. However, its role and mechanism in HCC are ill defined. In this article, we review the advanced insights into the progression of DDR1 in HCC, particularly the ligands and mechanisms in invasion and metastasis, which may open new avenues for the therapeutic utility of HCC.
Collapse
Affiliation(s)
- Linghong Wu
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xinhua Zhao
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Huan Ma
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Lili Zhang
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
| | - Xiaoan Li
- Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, Sichuan, People’s Republic of China
- Correspondence: Xiaoan Li, Department of Gastroenterology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 12 Changjia Lane, Jingzhong Street, Fucheng District, Mianyang, 621000, Sichuan, People’s Republic of China, Tel +86 816 224 3593 Email
| |
Collapse
|
44
|
MiR-29a Curbs Hepatocellular Carcinoma Incidence via Targeting of HIF-1α and ANGPT2. Int J Mol Sci 2022; 23:ijms23031636. [PMID: 35163556 PMCID: PMC8835722 DOI: 10.3390/ijms23031636] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/11/2022] Open
Abstract
A high-fat diet is responsible for hepatic fat accumulation that sustains chronic liver damage and increases the risks of steatosis and hepatocellular carcinoma (HCC). MicroRNA-29a (miR-29a), a key regulator of cellular behaviors, is present in anti-fibrosis and modulator tumorigenesis. However, the increased transparency of the correlation between miR-29a and the progression of human HCC is still further investigated. In this study, we predicted HIF-1α and ANGPT2 as regulators of HCC by the OncoMir cancer database and showed a strong positive correlation with HIF-1α and ANGPT2 gene expression in HCC patients. Mice fed the western diet (WD) while administered CCl4 for 25 weeks induced chronic liver damage and higher HCC incidence than without fed WD mice. HCC section staining revealed signaling upregulation in ki67, severe fibrosis, and steatosis in WD and CCl4 mice and detected Col3a1 gene expressions. HCC tissues significantly attenuated miR-29a but increased in HIF-1α, ANGPT2, Lox, Loxl2, and VEGFA expression. Luciferase activity analysis confirms that miR-29a specific binding 3′UTR of HIF-1α and ANGPT2 to repress expression. In summary, miR-29a control HIF-1α and ANGPT2 signaling in HCC formation. This study insight into a novel molecular pathway by which miR-29a targeting HIF-1α and ANGPT2 counteracts the incidence of HCC development.
Collapse
|
45
|
Chen W, Liang W, He Y, Liu C, Chen H, Lv P, Yao Y, Zhou H. Immune microenvironment-related gene mapping predicts immunochemotherapy response and prognosis in diffuse large B-cell lymphoma. Med Oncol 2022; 39:44. [PMID: 35092504 DOI: 10.1007/s12032-021-01642-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/23/2021] [Indexed: 01/01/2023]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is the most common subtype of non-Hodgkin's lymphoma (NHL). The R-CHOP immunochemotherapy regimen is the first-line treatment option for DLBCL patients and has greatly improved the prognosis of DLBCL, making it a curable disease. However, drug resistance or relapse is the main challenge for current DLBCL treatment. Studies have shown that the tumor microenvironment plays an important role in the onset, development, and responsiveness to drugs in DLBCL. Here, we used the CIBERSORT algorithm to resolve the composition of the immune microenvironment of 471 DLBCL patients from the GEO database. We found that activated memory CD4+ T cells and γδ T cells were significantly associated with immunochemotherapy response. Weighted gene co-expression networks (WGCNA) were constructed using differentially expressed genes from immunochemotherapy responders and non-responders. The module most associated with these two types of T cells was defined as hub module. Enrichment analysis of the hub module showed that baseline immune status was significantly stronger in responders than in non-responders. A protein-protein interaction (PPI) network was constructed for hub module to identify hub genes. After survival analysis, five prognosis-related genes (CD3G, CD3D, GNB4, FCHO2, GPR183) were identified and all these genes were significantly negatively associated with PD1. Using our own patient cohort, we validated the efficacy of CD3G and CD3D in predicting immunochemotherapy response. Our study showed that CD3G, CD3D, GNB4, FCHO2, and GPR183 are involved in the regulation of the immune microenvironment of DLBCL. They can be used as biomarkers for predicting immunochemotherapy response and potential therapeutic targets in DLBCL.
Collapse
Affiliation(s)
- Wanjun Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Weijie Liang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongjian He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chixiang Liu
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongtian Chen
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Piao Lv
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuan Yao
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huayou Zhou
- Department of Blood Transfusion, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
46
|
Li XY, Ma WN, Su LX, Shen Y, Zhang L, Shao Y, Wang D, Wang Z, Wen MZ, Yang XT. Association of Angiogenesis Gene Expression With Cancer Prognosis and Immunotherapy Efficacy. Front Cell Dev Biol 2022; 10:805507. [PMID: 35155426 PMCID: PMC8826089 DOI: 10.3389/fcell.2022.805507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Several new blood vessels are formed during the process of tumor development. These new blood vessels provide nutrients and water for tumour growth, while spreading tumour cells to distant areas and forming new metastases in different parts of the body. The available evidence suggests that tumour angiogenesis is closely associated with the tumour microenvironment and is regulated by a variety of pro-angiogenic factors and/or angiogenic inhibitors.Methods: In the present study, a comprehensive characterization of angiogenesis genes expression was performed in a pan-cancer analysis across the 33 human cancer types. Further, genetic data from several public databases were also used in the current study. An angiogenesis score was assigned to The Cancer Genome Atlas (TCGA) pan-cancer data, with one angiogenesis score as per sample for each tumour.Results: It was found that angiogenesis genes vary across cancer types, and are associated with a number of genomic and immunological features. Further, it was noted that macrophages and iTreg infiltration were generally higher in tumours with high angiogenesis scores, whereas lymphocytes and B cells showed the opposite trend. Notably, NK cells showed significantly different correlations among cancer types. Furthermore, results of the present study showed that a high angiogenesis score was associated with poor survival and aggressive types of cancer in most of the cancer types.Conclusion: In conclusion, the current study evidently showed that the expression of angiogenesis genes is a key feature of tumour biology that has a major impact on prognosis of patient with cancers.
Collapse
Affiliation(s)
- Xin-yu Li
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosurgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Ning Ma
- Department of Pediatrics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-xin Su
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuchen Shen
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liming Zhang
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhao Shao
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Deming Wang
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenfeng Wang
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ming-Zhe Wen
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-tao Yang
- Department of Interventional Therapy, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Xi-tao Yang,
| |
Collapse
|
47
|
Correlation between Immunohistochemical Markers in Hepatocellular Carcinoma Cells and In Vitro High-Throughput Drug Sensitivity Screening. Can J Gastroenterol Hepatol 2022; 2022:5969716. [PMID: 35127582 PMCID: PMC8808116 DOI: 10.1155/2022/5969716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
AIM This study analyzed the correlation between immunohistochemical markers in hepatocellular carcinoma cells and the results of in vitro high-throughput drug sensitivity screening, to provide a reference for individualized drug treatment in patients with liver cancer. METHODS Seventy-four patients with hepatocellular carcinoma were included in this study from December 2019 to June 2021, and their liver cancer cells were used for in vitro high-throughput drug sensitivity screening. According to the screening results, the patients were divided into relatively sensitive and insensitive groups, and the correlations between sensitivity and immunohistochemistry results were analyzed statistically. RESULTS Alpha-fetoprotein (AFP)-positive liver cancer cells were significantly more sensitive to gemcitabine than AFP-negative cells (χ 2 = 6.102, P=0.014). AFP was also positively correlated with sensitivity of liver cancer cells to three combined regimens containing oxaliplatin (L-OHP) and epirubicin (EPI) : L-OHP + EPI + irinotecan + 5-fluorouracil (5-FU), L-OHP + irinotecan + EPI, and L-OHP + EPI (χ 2 = 8.168, P=0.004, χ 2 = 5.705, P=0.017, and χ 2 = 8.275, P=0.004, respectively). CONCLUSION Gemcitabine and L-OHP + EPI + irinotecan + 5-FU, L-OHP + EPI, and L-OHP + irinotecan + EPI were more effective against AFP-positive compared with AFP-negative liver cancer cells according to in vitro high-throughput drug sensitivity screening. These results may guide the selection of personalized drug treatments for patients with advanced liver cancer in the future but still need further clinical studies to confirm.
Collapse
|
48
|
Yue C, Zhao T, Zhang S, Liu Y, Zheng G, Zhang Y. Comprehensive characterization of 11 prognostic alternative splicing events in ovarian cancer interacted with the immune microenvironment. Sci Rep 2022; 12:980. [PMID: 35046435 PMCID: PMC8770494 DOI: 10.1038/s41598-021-03836-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Alternative splicing (AS) events play a crucial role in the tumorigenesis and progression of cancer. Transcriptome data and Percent Spliced In (PSI) values of ovarian cancer patients were downloaded from TCGA database and TCGA SpliceSeq. Totally we identified 1472 AS events that were associated with survival of ovarian serous cystadenocarcinoma (OC) and exon skipping (ES) was the most important type. Univariate and multivariate Cox regression analysis were performed to identify survival-associated AS events and developed the prognostic model based on 11-AS events. The immune cells and different response to cytotoxic T lymphocyte associated antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) blockers in low-risk and high-risk group of OC patients were analyzed. Ten kinds of immune cells were found up-regulated in low-risk group. Activated B cell, natural killer T cell, natural killer cell and regulatory T cell were associated with survival of OC. The patients in low-risk group had good response to CTLA-4 and PD-1 blockers treatment. Moreover, a regulatory network was established according to the correlation between AS events and splicing factors (SFs). The present study provided valuable insights into the underlying mechanisms of OC. AS events that were correlated with the immune system might be potential therapeutic targets.
Collapse
Affiliation(s)
- Congbo Yue
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China
| | - Tianyi Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China
| | - Shoucai Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China
| | - Yingjie Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China
| | - Guixi Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China.
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong Province, 250012, People's Republic of China.
| |
Collapse
|
49
|
Witte HM, Gebauer N, Steinestel K. Mutational and immunologic Landscape in malignant Salivary Gland Tumors harbor the potential for novel therapeutic strategies. Crit Rev Oncol Hematol 2022; 170:103592. [PMID: 35026433 DOI: 10.1016/j.critrevonc.2022.103592] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 11/24/2021] [Accepted: 01/06/2022] [Indexed: 12/18/2022] Open
Abstract
Salivary gland carcinomas (SGC) are rare (3-6 % of all head and neck cancers) and show biological heterogeneity depending on the respective histological subtype. While complete surgical resection is the standard treatment for localized disease, chemotherapy or radiation therapy are frequently insufficient for the treatment of unresectable or metastasized SGC. Therefore, new therapeutic approaches such as molecularly targeted therapy or the application of immune checkpoint inhibition enhance the treatment repertoire. Accordingly, comprehensive analyses of the genomic landscape and the tumor-microenvironment (TME) are of crucial importance in order to optimize and individualize SGC treatment. This manuscript combines the current scientific knowledge of the composition of the mutational landscape and the TME in SGCs harboring the potential for novel (immune-) targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany; Department of Hematology and Oncology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany; Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany.
| | - Niklas Gebauer
- Department of Hematology and Oncology, University Hospital of Schleswig-Holstein, Campus Luebeck, 23538, Luebeck, Germany
| | - Konrad Steinestel
- Institute of Pathology and Molecular Pathology, Federal Armed Forces Hospital Ulm, Oberer Eselsberg 40, 89081, Ulm, Germany
| |
Collapse
|
50
|
Cretu I, Cretu B, Cirstoiu C, Cursaru A, Milicescu M, Bojinca M, Ionescu R. Rheumatological Adverse Events Following Immunotherapy for Cancer. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:94. [PMID: 35056402 PMCID: PMC8778975 DOI: 10.3390/medicina58010094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/02/2022] [Accepted: 01/06/2022] [Indexed: 06/14/2023]
Abstract
Background and Objectives: The occurrence of rheumatological side effects in a patient after receiving immunotherapy for cancer is becoming increasingly common. Oncologists often fail to diagnose and refer affected patients to rheumatologists. This paper presents the various rheumatological adverse events that occur after immunotherapy in patients as well as their treatment and evolution. Materials and Methods: A total of 36 patients were monitored between November 2018 and March 2020. The oncologist monitoring the immunotherapy-treated patients identified the occurrence of musculoskeletal side effects. The grading of toxicities was performed by both the oncologist and the rheumatologist using common terminology criteria for adverse events (CTCAE). Rheumatological treatment was administered, and for some patients, immunotherapy was discontinued. Results: The clinical presentations of the patients varied. Mild side effects (grade 1-2) were reported in a higher proportion than severe side effects (grade 3-5). Therefore, thirty-one patients had mild-to-moderate side effects, and five patients had severe side effects. Adverse reactions occurred, on average, 10 weeks after the initiation of immunotherapy; this indicated that the severity of the toxicity was dose dependent. Patients were treated with NSAIDs or prednisone, depending on the severity of the side effects, and for patients with severe manifestations, immunotherapy was discontinued. The remission of rheumatic manifestations varied depending on the grade of the manifestations. Conclusions: The clinical, biological, and ultrasound presentations of the patients with adverse events followed by cancer treatments differed from classic rheumatological manifestations. Thorough examinations of these patients by both oncologists and rheumatologists are needed in order to correctly diagnose and treat rheumatological adverse events. Multiple studies that include a larger number of participants are needed in order to better understand the pathogenesis and clinical evolution of these patients under different treatment conditions.
Collapse
Affiliation(s)
- Ioana Cretu
- Department Internal Medicine & Rheumatology, Carol Davila University of Medicine & Pharmacy, Dr Ion Cantacuzino Hospital, 917151 Bucharest, Romania; (I.C.); (M.M.); (M.B.)
| | - Bogdan Cretu
- Department Orthopaedics & Traumatology, Carol Davila University of Medicine & Pharmacy, University Emergency Hospital, 050098 Bucharest, Romania; (C.C.); (A.C.)
| | - Catalin Cirstoiu
- Department Orthopaedics & Traumatology, Carol Davila University of Medicine & Pharmacy, University Emergency Hospital, 050098 Bucharest, Romania; (C.C.); (A.C.)
| | - Adrian Cursaru
- Department Orthopaedics & Traumatology, Carol Davila University of Medicine & Pharmacy, University Emergency Hospital, 050098 Bucharest, Romania; (C.C.); (A.C.)
| | - Mihaela Milicescu
- Department Internal Medicine & Rheumatology, Carol Davila University of Medicine & Pharmacy, Dr Ion Cantacuzino Hospital, 917151 Bucharest, Romania; (I.C.); (M.M.); (M.B.)
| | - Mihai Bojinca
- Department Internal Medicine & Rheumatology, Carol Davila University of Medicine & Pharmacy, Dr Ion Cantacuzino Hospital, 917151 Bucharest, Romania; (I.C.); (M.M.); (M.B.)
| | - Ruxandra Ionescu
- Department Internal Medicine & Rheumatology, Carol Davila University of Medicine & Pharmacy, Sf Maria Clinical Hospital, 011172 Bucharest, Romania;
| |
Collapse
|