1
|
Garrido Martínez MT, Rodríguez Jorge M, García Giménez I, Guzmán Ramos MI, Grutzmancher Sáiz S, Aviñó Tarazona V. Toxicity of the FOLFOX-6 regimen, with or without 5-fluorouracil bolus, in metastatic colorectal cancer. FARMACIA HOSPITALARIA 2025; 49:154-159. [PMID: 39482145 DOI: 10.1016/j.farma.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/08/2024] [Accepted: 09/22/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Standard treatment of metastatic colorectal cancer includes oxaliplatin and 5-fluorouracil in continuous infusion. Although FOLFOX-6 is the reference combination, it is aggressive and has high toxicity. Variants such as the TTD regimen, which does not include folinic acid or 5-fluorouracil bolus, are used. This study evaluates the toxicity of FOLFOX-6 and TTD in first line treatment for metastatic colorectal cancer and its effectiveness. METHOD Retrospective observational study with patients who started treatment with FOLFOX-6 and TTD, for three years. Demographic and clinical data were collected (age, sex, chronic pathologies, molecular profile, laterality, ECOG classification and stage), as well as treatment variables (previous adjuvant chemotherapy, intentionality, number of cycles, duration and pharmacogenetic aspects) and toxicity. Objective response rate and progression-free survival were calculated. RESULTS The study included 71 patients, 35 treated with FOLFOX-6 and 36 with TTD. Both groups showed similar overall toxicity profiles. FOLFOX-6 had a higher incidence of neutropenia (46% vs. 8%; p < 0.01) and mucositis (51% vs. 22%; p < 0.013). In addition, there were more treatment delays (40% vs. 11%; p < 0.05) and 5-fluorouracil dose reductions (22% vs. 14%; p < 0.05) in the FOLFOX-6 group. Deaths due to toxicity were only recorded in the FOLFOX-6 group. Effectiveness was similar in both groups. CONCLUSIONS The TTD regimen could be a beneficial first-line option for metastatic colorectal cancer, with lower toxicity and effectiveness comparable to FOLFOX-6. It is a safe alternative for elderly or frail patients, suitable for reduced-dose 5-fluorouracil regimen with oxaliplatin.
Collapse
Affiliation(s)
| | - María Rodríguez Jorge
- Servicio de Farmacia Hospitalaria, Hospital Universitario Juan Ramón Jiménez, Huelva, España.
| | - Ignacio García Giménez
- Servicio de Farmacia Hospitalaria, Hospital Universitario Juan Ramón Jiménez, Huelva, España
| | | | | | | |
Collapse
|
2
|
Suleiman H, Emerson A, Wilson PM, Mulligan KA, Ladner RD, LaBonte MJ. Harnessing nucleotide metabolism and immunity in cancer: a tumour microenvironment perspective. FEBS J 2025; 292:2155-2172. [PMID: 39308084 PMCID: PMC12062787 DOI: 10.1111/febs.17278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/06/2024] [Accepted: 09/09/2024] [Indexed: 05/11/2025]
Abstract
The tumour microenvironment (TME) is a dynamic nexus where cancer cell metabolism and the immune system intricately converge, with nucleotide metabolism (NM) playing a pivotal role. This review explores the critical function of NM in cancer cell proliferation and its profound influence on the TME and immune landscape. NM is essential for DNA and RNA synthesis and is markedly upregulated in cancer cells to meet the demands of rapid growth. This metabolic rewiring fuels cancer progression, but also shapes the TME, impacting the function and viability of immune cells. The altered nucleotide milieu in the TME can suppress immune response, aiding cancer cell evasion from immune surveillance. Drug discoveries in the field of NM have revealed different therapeutic strategies, including inhibitors of nucleotide synthesis and drugs targeting salvage pathways, which are discussed thoroughly in this review. Furthermore, the emerging strategy of combining NM-targeted therapies with immunotherapies is emphasised, particularly their effect on sensitising tumours to immune checkpoint inhibitors and enhancing overall treatment efficacy. The Human Genome Project paved the way for personalised medicine, countering the established 'one size fits all' approach to cancer treatment. Advances in understanding the TME and NM have spurred interest in personalised therapeutic strategies. This review highlights the potential of leveraging individual tumour metabolic profiles to guide treatment selection, aiming to optimise efficacy and minimise adverse effects. The strategic importance of targeting NM in cancer therapy and its synergistic potential with immunotherapies offers a path towards more effective and personalised cancer treatments.
Collapse
Affiliation(s)
- Hadil Suleiman
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Alexandra Emerson
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | | | | | - Robert D. Ladner
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
- CV6 Therapeutics (NI) LtdBelfastUK
| | - Melissa J. LaBonte
- Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastUK
| |
Collapse
|
3
|
Garrido Martínez MT, Rodríguez Jorge M, García Giménez I, Guzmán Ramos MI, Grutzmancher Sáiz S, Aviñó Tarazona V. [Translated article] Toxicity of the FOLFOX-6 regimen, with or without 5-fluorouracil bolus, in metastatic colorectal cancer. FARMACIA HOSPITALARIA 2025; 49:T154-T159. [PMID: 39875275 DOI: 10.1016/j.farma.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/08/2024] [Accepted: 09/22/2024] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVE Standard treatment of metastatic colorectal cancer includes oxaliplatin and 5-fluorouracil in continuous infusion. Although FOLFOX-6 is the reference combination, it is aggressive and has high toxicity. Variants such as the TTD regimen, which does not include folinic acid or 5-fluorouracil bolus, are used. This study evaluates the toxicity of FOLFOX-6 and TTD in first line treatment for metastatic colorectal cancer and its effectiveness. METHOD Retrospective observational study with patients who started treatment with FOLFOX-6 and TTD, for 3 years. Demographic and clinical data were collected (age, sex, chronic pathologies, molecular profile, laterality, Eastern Cooperative Oncology Group classification, and stage), as well as treatment variables (previous adjuvant chemotherapy, intentionality, number of cycles, duration, and pharmacogenetic aspects) and toxicity. Objective response rate and progression-free survival were calculated. RESULTS The study included 71 patients, 35 treated with FOLFOX-6, and 36 with TTD. Both groups showed similar overall toxicity profiles. FOLFOX-6 had a higher incidence of neutropenia (46% vs 8%; P < .01) and mucositis (51% vs 22%; P < .013). In addition, there were more treatment delays (40% vs 11%; P < .05) and 5-fluorouracil dose reductions (22% vs 14%; P < .05) in the FOLFOX-6 group. Deaths due to toxicity were only recorded in the FOLFOX-6 group. Effectiveness was similar in both groups. CONCLUSIONS The TTD regimen could be a beneficial first-line option for metastatic colorectal cancer, with lower toxicity and effectiveness comparable to FOLFOX-6. It is a safe alternative for elderly or frail patients, suitable for reduced-dose 5-fluorouracil regimen with oxaliplatin.
Collapse
Affiliation(s)
| | - María Rodríguez Jorge
- Servicio de Farmacia Hospitalaria, Hospital Universitario Juan Ramón Jiménez, Huelva, Spain
| | - Ignacio García Giménez
- Servicio de Farmacia Hospitalaria, Hospital Universitario Juan Ramón Jiménez, Huelva, Spain
| | | | | | | |
Collapse
|
4
|
Papakonstantinou M, Fantakis A, Torzilli G, Donadon M, Chatzikomnitsa P, Giakoustidis D, Papadopoulos VN, Giakoustidis A. A Systematic Review of Disappearing Colorectal Liver Metastases: Resection or No Resection? J Clin Med 2025; 14:1147. [PMID: 40004679 PMCID: PMC11856073 DOI: 10.3390/jcm14041147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Colorectal cancer is the second most common type of cancer and a leading cause of cancer-related deaths worldwide. Approximately 15% of the patients with colorectal cancer will already have liver metastases (CRLMs) at diagnosis. Luckily, the advances in chemotherapy regimens during the past few decades have led to increased rates of disease regression that could even render an originally unresectable disease resectable. In certain patients with CRLMs, the hepatic lesions are missing on preoperative imaging after neoadjuvant chemotherapy. These patients can undergo surgery with or without resection of the sites of the disappearing liver metastases (DLMs). In this systematic review, we assess the recurrence rate of the DLMs that were left unresected as well as the complete pathologic response of those resected. Methods: A literature search was conducted in PubMed for studies including patients with CRLMs who received neoadjuvant chemotherapy and had DLMs in preoperative imaging. Two independent reviewers completed the search according to the PRISMA checklist. Results: Three hundred and twenty-six patients with 1134 DLMs were included in our review. A total of 47 out of 480 DLMs (72.29%) that were removed had viable tumor cells in postoperative histology. One hundred and forty-five tumors could not be identified intraoperatively and were removed based on previous imaging, with thirty (20.69%) of them presenting viable cancer cells. Four hundred and sixty-five lesions could not be identified and were left in place. Of them, 152 (32.69%) developed local recurrence within 5 years. Of note, 34 DLMs could not be categorized as viable or non-viable tumors. Finally, DLMs that were identifiable intraoperatively had a higher possibility of viable tumors compared to non-identifiable ones (72.29% vs. 20.69%, respectively). Conclusions: Disappearing liver metastases that are left unresected have an increased possibility of recurrence. Patients receiving neoadjuvant treatment for CRLMs may have better survival chances after resecting all the DLM sites, either identifiable intraoperatively or not.
Collapse
Affiliation(s)
- Menelaos Papakonstantinou
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| | - Antonios Fantakis
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| | - Guido Torzilli
- Department of Surgery, Division of Hepatobiliary Surgery & General Surgery, Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Matteo Donadon
- Surgical Oncology Program, University Maggiore Hospital, University of Piemonte Orientale, 28100 Novara, Italy;
| | - Paraskevi Chatzikomnitsa
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| | - Dimitrios Giakoustidis
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| | - Vasileios N. Papadopoulos
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| | - Alexandros Giakoustidis
- Aristotle University Surgery Department, Papageorgiou Hospital, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (M.P.); (A.F.); (P.C.); (D.G.); (V.N.P.)
| |
Collapse
|
5
|
Huo M, Gao Z, Wang G, Hou Z, Zheng J. Huaier promotes sensitivity of colorectal cancer to oxaliplatin by inhibiting METTL3 to regulate the Wnt/β‑catenin signaling pathway. Oncol Rep 2025; 53:7. [PMID: 39513580 PMCID: PMC11574703 DOI: 10.3892/or.2024.8840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/24/2024] [Indexed: 11/15/2024] Open
Abstract
Colorectal cancer (CRC) ranks fifth in terms of incidence rate and mortality among malignant tumors in China. Oxaliplatin (OXA) is a first‑line drug for the clinical treatment of CRC, but its antitumor effect is limited because of the development of drug resistance. The present study aimed to investigate whether the traditional Chinese medicine Huaier can regulate the Wnt/β‑catenin signaling pathway by affecting the expression of METTL3, thereby promoting the sensitivity of HCT‑8/L cells to OXA. The expression of METTL3 was analyzed based on the UCSC Xena and Gene Expression Omnibus databases. Silent METTL3 and overexpression METTL3 models were constructed, and Cell Counting Kit‑8 and flow cytometry were used to detect the effects of Huaier on the viability and apoptosis of HCT‑8/L cells. Western blotting, reverse transcription‑quantitative PCR, nuclear cytoplasmic separation and immunofluorescence were used to detect the effects of Huaier on the expression of METTL3, Pgp, Wnt/β‑catenin signaling pathway‑related proteins, apoptosis‑related proteins and related mRNA. The results demonstrated that patients with high expression levels of METTL3 had a shorter overall survival period. The expression level of METTL3 significantly increased in drug‑resistant CRC cells. Silencing METTL3 promoted apoptosis of CRC cells and increased their sensitivity to OXA by inhibiting the Wnt/β‑catenin signaling pathway. Huaier downregulated the expression of METTL3, thereby promoting apoptosis of drug‑resistant CRC cells and increasing their sensitivity to OXA by inhibiting the Wnt/β‑catenin signaling pathway.
Collapse
Affiliation(s)
- Mingyi Huo
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhixu Gao
- Department of Traditional Chinese Medicine, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Guizhen Wang
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Zhiping Hou
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Jining Zheng
- Department of Pathophysiology, Chengde Medical University, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
6
|
Yousef M, Yousef A, Chowdhury S, Fanaeian MM, Knafl M, Peterson J, Zeineddine M, Alfaro K, Zeineddine F, Goldstein D, Hornstein N, Dasari A, Huey R, Johnson B, Higbie V, Bent A, Kee B, Lee M, Morelli MP, Morris VK, Halperin D, Overman MJ, Parseghian C, Vilar E, Wolff R, Raghav KP, White MG, Uppal A, Sun R, Wang W, Kopetz S, Willis J, Shen JP. Molecular, Socioeconomic, and Clinical Factors Affecting Racial and Ethnic Disparities in Colorectal Cancer Survival. JAMA Oncol 2024; 10:1519-1529. [PMID: 39264607 PMCID: PMC11393757 DOI: 10.1001/jamaoncol.2024.3666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/05/2024] [Indexed: 09/13/2024]
Abstract
Importance Disparity in overall survival (OS) and differences in the frequency of driver gene variants by race and ethnicity have been separately observed in patients with colorectal cancer; however, how these differences contribute to survival disparity is unknown. Objective To quantify the association of molecular, socioeconomic, and clinical covariates with racial and ethnic disparities in overall survival among patients with colorectal cancer. Design, Setting, and Participants This single-center cohort study was conducted at a tertiary-level cancer center using relevant data on all patients diagnosed with colorectal cancer from January 1, 1973, to March 1, 2023. The relative contribution of variables to the disparity was determined using mediation analysis with sequential multivariate Cox regression models. Main Outcome OS, from diagnosis date and from start of first-line chemotherapy. Results The study population of 47 178 patients (median [IQR] age, 57.0 [49-66] years; 20 465 [43.4%] females and 26 713 [56.6%] males; 3.0% Asian, 8.7% Black, 8.8% Hispanic, and 79.4% White individuals) had a median (IQR) follow-up from initial diagnosis of 124 (174) months and OS of 55 (145) months. Compared with White patients, Black patients had worse OS (hazard ratio [HR], 1.16; 95% CI, 1.09-1.24; P <.001), whereas Asian and Hispanic patients had better OS (HR, 0.66; 95% CI, 0.59-0.74; P <.001; and 0.86; 95% CI, 0.81-0.92; P <.001, respectively). When restricted to patients with metastatic disease, the greatest disparity was between Black patients compared with White patients (HR, 1.2; 95% CI, 1.06-1.37; P <.001). Evaluating changes in OS disparity over 20 years showed disparity decreasing among Asian, Hispanic, and White patients, but increasing between Black patients and White patients (HRs, 1.18; 95% CI, 1.07-1.31 for 2008-2012; 1.24, 95% CI, 1.08-1.42 for 2013-2017; and 1.50; 95% CI, 1.20-1.87 for 2018-2023). Survival outcomes for first-line chemotherapy were worse for Black patients compared with White patients (median OS, 18 vs 26 months; HR, 1.30; 95% CI, 1.01-1.70). Among 7628 patients who had clinical molecular testing, APC, KRAS, and PIK3CA showed higher variant frequency in Black patients (false discovery rate [FDR], 0.01; < 0.001; and 0.01, respectively), whereas BRAF and KIT were higher among White patients (FDR, 0.001 and 0.01). Mediation analysis identified neighborhood socioeconomic status as the greatest contributor to OS disparity (29%), followed by molecular characteristics (microsatellite instability status, KRAS variation and BRAF variation, 10%), and tumor sidedness (9%). Conclusions This single-center cohort study identified substantial OS disparity and differing frequencies of driver gene variations by race and ethnicity. Socioeconomic status had the largest contribution but accounted for less than one-third of the disparity, with substantial contribution from tumor molecular features. Further study of the associations of genetic ancestry and the molecular pathogenesis of colorectal cancer with chemotherapy response is needed.
Collapse
Affiliation(s)
- Mahmoud Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Abdelrahman Yousef
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Saikat Chowdhury
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad M. Fanaeian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mark Knafl
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer Peterson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Mohammad Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kristin Alfaro
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Fadl Zeineddine
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | | | - Nicholas Hornstein
- Department of General Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Arvind Dasari
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Ryan Huey
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Benny Johnson
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Victoria Higbie
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Alisha Bent
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Bryan Kee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael Lee
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Maria Pia Morelli
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Daniel Halperin
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael J. Overman
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Christine Parseghian
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, University of Texas MD Anderson Cancer Center, Houston
| | - Robert Wolff
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Kanwal P. Raghav
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Michael G. White
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Abhineet Uppal
- Department of Colon and Rectal Surgery, University of Texas MD Anderson Cancer Center, Houston
| | - Ryan Sun
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - Jason Willis
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| | - John Paul Shen
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
7
|
Anselmino LE, Malizia F, Avila A, Cesatti Laluce N, Mamberto M, Zanotti LC, Farré C, Sauzeau V, Menacho Márquez M. Overcoming Therapy Resistance in Colorectal Cancer: Targeting the Rac1 Signaling Pathway as a Potential Therapeutic Approach. Cells 2024; 13:1776. [PMID: 39513883 PMCID: PMC11545287 DOI: 10.3390/cells13211776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/10/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Colorectal cancer (CRC) is the third most commonly diagnosed type of cancer worldwide and is responsible for numerous deaths. 5-fluorouracil (5-FU) is an effective chemotherapy drug commonly used in the treatment of CRC, either as monotherapy or in combination with other drugs. However, half of CRC cases are resistant to 5-FU-based therapies. To contribute to the understanding of the mechanisms underlying CRC resistance or recurrence after 5-FU-based therapies, we performed a comprehensive study integrating in silico, in vitro, and in vivo approaches. We identified differentially expressed genes and enrichment of pathways associated with recurrence after 5-FU-based therapies. Using these bioinformatics data as a starting point, we selected a group of drugs that restored 5-FU sensitivity to 5-FU resistant cells. Interestingly, treatment with the novel Rac1 inhibitor, 1A-116, reversed morphological changes associated with 5-FU resistance.. Moreover, our in vivo studies have shown that 1A-116 affected tumor growth and the development of metastasis. All our data allowed us to postulate that targeting Rac1 represents a promising avenue for the development of new treatments for patients with CRC resistant to 5-FU-based therapies.
Collapse
Affiliation(s)
- Luciano E. Anselmino
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Florencia Malizia
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Aylén Avila
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Nahuel Cesatti Laluce
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Macarena Mamberto
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Lucía C. Zanotti
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Cecilia Farré
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| | - Vincent Sauzeau
- Institut du Thorax, Inserm, CNRS, Université de Nantes, 44000 Nantes, France;
| | - Mauricio Menacho Márquez
- Instituto de Inmunología Clínica y Experimental de Rosario (IDICER, CONICET-UNR), Facultad de Ciencias Médicas (UNR), Rosario 2000, Argentina; (L.E.A.); (F.M.); (N.C.L.); (M.M.); (L.C.Z.); (C.F.)
- Instituto de Inmunología Clínica y Experimental, CONICET, Rosario 2000, Argentina
- Centro de Investigación y Producción de Reactivos Biológicos (CIPReB), Facultad de Ciencias Médicas (UNR), Suipacha 660, Rosario 2000, Argentina;
- Centro de Investigación del Cáncer de Rosario (CIC-R), Red de Investigación del Cáncer de Rosario (RICaR), Rosario 2000, Argentina
| |
Collapse
|
8
|
Chen JK, Merrick KA, Kong YW, Izrael-Tomasevic A, Eng G, Handly ED, Patterson JC, Cannell IG, Suarez-Lopez L, Hosios AM, Dinh A, Kirkpatrick DS, Yu K, Rose CM, Hernandez JM, Hwangbo H, Palmer AC, Vander Heiden MG, Yilmaz ÖH, Yaffe MB. An RNA damage response network mediates the lethality of 5-FU in colorectal cancer. Cell Rep Med 2024; 5:101778. [PMID: 39378883 PMCID: PMC11514606 DOI: 10.1016/j.xcrm.2024.101778] [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: 05/31/2024] [Revised: 07/15/2024] [Accepted: 09/16/2024] [Indexed: 10/10/2024]
Abstract
5-fluorouracil (5-FU), a major anti-cancer therapeutic, is believed to function primarily by inhibiting thymidylate synthase, depleting deoxythymidine triphosphate (dTTP), and causing DNA damage. Here, we show that clinical combinations of 5-FU with oxaliplatin or irinotecan show no synergy in human colorectal cancer (CRC) trials and sub-additive killing in CRC cell lines. Using selective 5-FU metabolites, phospho- and ubiquitin proteomics, and primary human CRC organoids, we demonstrate that 5-FU-mediated CRC cell killing primarily involves an RNA damage response during ribosome biogenesis, causing lysosomal degradation of damaged rRNAs and proteasomal degradation of ubiquitinated ribosomal proteins. Tumor types clinically responsive to 5-FU treatment show upregulated rRNA biogenesis while 5-FU clinically non-responsive tumor types do not, instead showing greater sensitivity to 5-FU's DNA damage effects. Finally, we show that treatments upregulating ribosome biogenesis, including KDM2A inhibition, promote RNA-dependent cell killing by 5-FU, demonstrating the potential for combinatorial targeting of this ribosomal RNA damage response for improved cancer therapy.
Collapse
Affiliation(s)
- Jung-Kuei Chen
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Karl A Merrick
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yi Wen Kong
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - George Eng
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Erika D Handly
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jesse C Patterson
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ian G Cannell
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lucia Suarez-Lopez
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aaron M Hosios
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anh Dinh
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Kebing Yu
- Genentech Biotechnology company, South San Francisco, CA 94080, USA
| | | | - Jonathan M Hernandez
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haeun Hwangbo
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pharmacology, Computational Medicine Program, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam C Palmer
- Department of Pharmacology, Computational Medicine Program, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew G Vander Heiden
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Ömer H Yilmaz
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B Yaffe
- Center for Precision Cancer Medicine, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Surgery, Beth Israel Medical Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
9
|
Singhal Y, Pingoliya SK, Saji S, Gaurav RP. Comparison of Efficacy and Safety of Prophylactic Use of Metoclopramide and Haloperidol on Morphine-induced Nausea and Vomiting in Cancer Patients: A Comparative, Randomised, Prospective Study. Indian J Palliat Care 2024; 30:375-379. [PMID: 39650588 PMCID: PMC11618674 DOI: 10.25259/ijpc_141_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/12/2024] [Indexed: 12/11/2024] Open
Abstract
Objectives Morphine is the cornerstone of pain management in a palliative care setting. Nausea with or without vomiting usually occurs when patients are initiated on morphine for the 1st time or when the dose is substantially increased. Materials and Methods A total of 90 patients fulfilling the inclusion criteria were randomly allocated into two groups of 45 each. Group M received a tablet of metoclopramide 10 mg orally 3 times a day; Group H received a tablet of haloperidol (2.5 mg) orally at night. All the patients were provided NCI CTCAE V4.3 NAUSEA AND VOMITING SCALE and asked to mark their response from day 1 to day 7. At the follow-up visit on the 7th day, the form was collected, and data were analysed. Results In Group M and Group H, the mean nausea score was between 1 and 2 and the difference was statistically insignificant. In Group M, the maximum vomiting score was 1.28 on day 5, while in Group H, the maximum score was 2 on day 5. The difference between the two groups was statistically significant. Conclusion Metoclopramide and haloperidol are equally efficacious in preventing nausea, but metoclopramide was found to be more effective with lesser side effects than haloperidol for morphine-induced vomiting in cancer patients when used prophylactically.
Collapse
Affiliation(s)
- Yogendra Singhal
- Department of Palliative Medicine, Sawai Man Singh Medical College, Jaipur, Rajasthan, India
| | | | - Sreeharsh Saji
- Department of Palliative Medicine, Sawai Man Singh Medical College, Jaipur, Rajasthan, India
| | - R. Pavan Gaurav
- Department of Palliative Medicine, Sawai Man Singh Medical College, Jaipur, Rajasthan, India
| |
Collapse
|
10
|
Rauwerdink P, van de Vlasakker VCJ, Wassenaar ECE, Rovers KP, Los M, Herbschleb KH, Creemers GJM, Thijs AMJ, Raicu MG, Huysentruyt CJR, van der Hoeven EJRJ, Nederend J, Peeters RYM, Deenen MJ, Elias SG, Fijneman RJA, Constantinides A, Kranenburg O, Burger PWA, Nienhuijs SW, Wiezer RJ, Lurvink RJ, de Hingh IHJT, Boerma D. First-line palliative systemic therapy alternated with oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy for unresectable colorectal peritoneal metastases: A single-arm phase II trial (CRC-PIPAC-II). EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108487. [PMID: 38905732 DOI: 10.1016/j.ejso.2024.108487] [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: 03/10/2024] [Revised: 05/22/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Palliative systemic therapy alternated with electrostatic precipitation oxaliplatin-based pressurized intraperitoneal aerosol chemotherapy (ePIPAC) has never been prospectively investigated in patients with unresectable colorectal peritoneal metastases (CPM). The CRC-PIPAC-II study aimed to assess safety, feasibility and efficacy of such bidirectional therapy. METHODS This two-center, single-arm, phase II trial enrolled chemotherapy-naïve patients to undergo three treatment cycles, consisting of systemic therapy (CAPOX, FOLFOX, FOLFIRI, or FOLFOXIRI, all with bevacizumab) and oxaliplatin-based ePIPAC (92 mg/m2) with intravenous leucovorin (20 mg/m2) and 5-fluorouracil (400 mg/m2). Primary outcome were major treatment-related adverse events. Secondary outcomes included minor events, tumor response, progression-free survival (PFS) and overall survival (OS). RESULTS Twenty patients completed 52 treatment cycles. Fifteen major events occurred in 7 patients (35 %): 5 events (33 %) related to systemic therapy; 5 (33 %) related to ePIPAC; and 5 (33 %) were biochemical events. No treatment-related deaths occurred. All patients experienced minor events, mostly abdominal pain, nausea and peripheral sensory neuropathy. After treatment, radiological, pathological, cytological, and biochemical response was observed in 0 %, 88 %, 38 %, and 31 % of patients respectively. Curative surgery was achieved in one patient. Median PFS was 10.0 months (95 % confidence interval [CI] 8.0-13.0) and median OS was 17.5 months (95 % CI 13.0-not reached). CONCLUSIONS Combining palliative systemic therapy with oxaliplatin-based ePIPAC in patients with unresectable CPM was feasible and showed an acceptable safety profile. Treatment-induced response and survival are promising, yet further research is required to determine the additional value of ePIPAC to systemic therapy.
Collapse
Affiliation(s)
| | | | | | - Koen P Rovers
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Maartje Los
- Department of Medical Oncology, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Karin H Herbschleb
- Department of Medical Oncology, St. Antonius Hospital, Nieuwegein, Netherlands
| | | | | | - Mihaela G Raicu
- Department of Pathology DNA, St. Antonius Hospital, Nieuwegein, Netherlands
| | | | | | - Joost Nederend
- Department of Radiology, Catharina Hospital, Eindhoven, Netherlands
| | - Rifka Y M Peeters
- Department of Clinical Pharmacy, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Maarten J Deenen
- Department of Clinical Pharmacy, Catharina Hospital, Eindhoven, Netherlands; Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, Netherlands
| | - Sjoerd G Elias
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Alexander Constantinides
- Lab Translational Oncology, Division Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Onno Kranenburg
- Lab Translational Oncology, Division Imaging and Cancer, University Medical Center Utrecht, Utrecht, Netherlands
| | - Pim W A Burger
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | | | - René J Wiezer
- Department of Surgery, St. Antonius Hospital, Nieuwegein, Netherlands
| | - Robin J Lurvink
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, Netherlands; Department of Epidemiology, School for Oncology and Developmental Biology, GROW, Maastricht, Netherlands.
| | - Djamila Boerma
- Department of Surgery, St. Antonius Hospital, Nieuwegein, Netherlands.
| |
Collapse
|
11
|
Keshavarzi F, Salari N, Jambarsang S, Mohammad Tabatabaei S, Shahsavari S, Fournier AJ. Overall survival with non-proportional hazards in first-line treatment for patients with metastatic colorectal cancer: Systematic review and network meta-analysis. Heliyon 2024; 10:e36464. [PMID: 39253267 PMCID: PMC11381762 DOI: 10.1016/j.heliyon.2024.e36464] [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: 11/30/2023] [Revised: 08/15/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
This study aimed to identify the most effective first-line treatment for patients with metastatic colorectal cancer based on overall survival, identify the most commonly used treatment, and generate a meaningful ranking among all available treatments based on their relative effectiveness. Researchers used the ANOVA parametrization method to fit the second-order fractional polynomial network meta-analysis with a random-effect model. Using a non-proportional hazards network meta-analysis, 46 treatments were compared by considering a combination of direct and indirect evidence extracted from clinical trial studies. Included in the review were 46 trials involving 21350 patients. Between January 2000 and January 2023, researchers conducted a thorough search through Embase, PubMed/Medline, and Scopus. To undertake a secondary analysis of this data, we recreate individual patient data from published Kaplan-Meier (K-M) survival curves and assess the accuracy of that reconstruction. A random-effects model was used to evaluate the pooled overall survival and hazard ratio with a 95 percent confidence interval. The predicted survival curves for the network meta-analysis showed that GOLFIG and FOLFOX + Cetuximab treatments have higher survival, respectively. Our results provide moderate quality evidence and comparative effective estimates for various available first-line treatments for metastasis colorectal cancer based on network meta-analysis.
Collapse
Affiliation(s)
- Fatemeh Keshavarzi
- Department of Biostatistics, Faculty of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Nader Salari
- Department of Biostatistics, Faculty of Health, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sara Jambarsang
- Department of Bio-Statistics and Epidemiology, Shahid Sadoughi University of Medical Science, Yazd, Iran
| | - Seyyed Mohammad Tabatabaei
- Department of Medical Informatics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodeh Shahsavari
- Department of Health Information Management, School of Allied Medical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | |
Collapse
|
12
|
Gaetani RS, Ladin K, Abelson JS. Journey through the Decades: The Evolution in Treatment and Shared Decision Making for Locally Advanced Rectal Cancer. Cancers (Basel) 2024; 16:2807. [PMID: 39199579 PMCID: PMC11353159 DOI: 10.3390/cancers16162807] [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: 07/12/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/01/2024] Open
Abstract
The management of locally advanced rectal cancer has undergone significant transformations over the decades and optimal treatment approaches continue to evolve. There have been numerous advances in surgery, chemotherapy, and radiation therapy from the first description of the abdominoperineal resection in 1908, timing of chemotherapy and radiation therapy in the late 20th and early 21st century, and most recently, the introduction of organ preservation or nonoperative management in 2004. Alongside these advancements, the concept of shared decision making in medicine has evolved, prompting a focus on patient-centered care. This evolution in practice has been fueled by a growing recognition of the importance of patient autonomy and the alignment of treatment options with patients' values and preferences. With the growing number of possible treatment options, variability in patient counseling exists, highlighting the need for a standardized approach to shared decision making in locally advanced rectal cancer. This narrative review will describe the evolution of treatment options of locally advanced rectal cancer as well as the concept of shared decision making and decision aids, and will introduce a decision aid for patients with locally advanced rectal cancer who have achieved a complete clinical response and are eligible for watch and wait.
Collapse
Affiliation(s)
- Racquel S. Gaetani
- Department of Colon and Rectal Surgery, Lahey Hospital and Medical Center, Burlington, MA 01805, USA;
| | - Keren Ladin
- Department of Community Health, Tufts University, Medford, MA 02155, USA
| | - Jonathan S. Abelson
- Department of Colon and Rectal Surgery, Lahey Hospital and Medical Center, Burlington, MA 01805, USA;
| |
Collapse
|
13
|
He Y, Li F, Zhang Y, Zhu X, Lin Z, Li L, Nawaz S, Kulyar MFEA, Iqbal M, Li J. Pediococcus pentosaceus PP34 Ameliorates 5-Fluorouracil-Induced Intestinal Mucositis via Inhibiting Oxidative Stress and Restoring the Gut Microbiota. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10324-1. [PMID: 39046671 DOI: 10.1007/s12602-024-10324-1] [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] [Accepted: 07/08/2024] [Indexed: 07/25/2024]
Abstract
Chemotherapy-induced intestinal mucositis based on 5-fluorouracil (5-FU) slows down the progress of cancer treatment and causes significant suffering to patients. Pediococcus pentosaceus (P. pentosaceus), as a type of LAB, has a range of probiotic properties, including antioxidant, immune benefits, and cholesterol-lowering effects, which are attracting increasing attention. However, studies on the protective effect of P. pentosaceus against chemotherapeutic-induced intestinal mucositis caused by 5-FU remain unclear. Therefore, this study aimed to investigate the potential relieving effects of P. pentosaceus PP34 on 5-FU-induced intestinal mucositis and its mechanism. In the present study, a P. pentosaceus PP34 solution (2 × 109 CFU/mL) was administered daily by gavage followed by intraperitoneal injection of 5-FU to model intestinal mucositis. The body weight, serum biochemical indices, jejunal pathological organization, and expression levels of inflammatory cytokines in the jejunum were examined. The results indicated that the mice induced with 5-FU developed typical intestinal mucositis symptoms and histopathological changes with intense inflammatory and oxidative responses. Moreover, the gut microbiota was disturbed, while PP34 effectively decreased the oxidative reactions and the expression levels of inflammatory mediators and regulated the gut microbiota in 5-FU-exposed mice. Taken together, the study indicated that P. pentosaceus PP34 ameliorates 5-Fluorouracil-induced intestinal mucositis via inhibiting oxidative stress and restoring the gut microbiota.
Collapse
Affiliation(s)
- Yuanyuan He
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Feiran Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiaohui Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Zhengrong Lin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Linxiao Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | | | - Mudassar Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jiakui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
- College of Animals Husbandry and Veterinary Medicine, Tibet Agricultural and Animal Husbandry University, Linzhi, Tibet, 860000, People's Republic of China.
| |
Collapse
|
14
|
Zhan Y, Cheng X, Mei P, Tan S, Feng W, Jiang H. Safety of first-line systemic therapy in patients with metastatic colorectal cancer: a network meta-analysis of randomized controlled trials. BMC Cancer 2024; 24:893. [PMID: 39048944 PMCID: PMC11270896 DOI: 10.1186/s12885-024-12662-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE To evaluate the safety of first-line systemic therapy for metastatic colorectal cancer through network meta-analysis. METHODS The literature from PubMed, Embase, Web of Science, and Cochrane Library databases was searched from the inception of the databases to August 15, 2023, and strict inclusion and exclusion criteria were applied to screen studies. The Cochrane Bias Risk Assessment Tool (RoB 2.0) was used to evaluate the quality of the included literature. Network meta-analysis was conducted using Stata 15.0 and R4.3.1 software to compare the incidence of adverse events (AEs) among different treatment regimens. RESULTS A total of 53 randomized controlled trials, involving 17,351 patients with metastatic colorectal cancer (mCRC), were ultimately included, encompassing 29 different therapeutic approaches. According to SUCRA rankings, the CAPOX regimen is most likely to rank first in terms of safety, while the FOLFOXIRI + panitumumab regimen is most likely to rank last. In terms of specific AEs, the CAPOX regimen, whether used alone or in combination with targeted drugs (bevacizumab and cetuximab), is associated with a reduced risk of neutropenia and febrile neutropenia, as well as an increased risk of thrombocytopenia and diarrhea. The FOLFOX regimen, with or without bevacizumab, is linked to an increased risk of neutropenia and peripheral sensory neuropathy. The FOLFIRI/CAPIRI + bevacizumab regimen is associated with a reduced risk of peripheral sensory neuropathy. S-1 and S-1 + oxaliplatin are well-tolerated in terms of gastrointestinal reactions. The FOLFOXIRI regimen, whether used alone or in combination with targeted drugs, is associated with various AEs. CONCLUSION In summary, the CAPOX regimen may be the safest option among the first-line systemic treatment regimens for mCRC patients, while the FOLFOXIRI + panitumumab regimen may be associated with a higher incidence of grade 3 or higher AEs.
Collapse
Affiliation(s)
- Yanrong Zhan
- Rudong People's Hospital / Affiliated Rudong Hospital of Xinglin College, Nantong University, Nantong, Jiangsu, 226400, China.
| | - Xianwen Cheng
- Ankang Hospital of Traditional Chinese Medicine, Ankang, Shaanxi, 725000, China
| | - Pingping Mei
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Shufa Tan
- Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China
| | - Wenzhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China.
| | - Hua Jiang
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, 712000, China
| |
Collapse
|
15
|
Heczko L, Liška V, Vyčítal O, Fiala O, Šůsová S, Hlaváč V, Souček P. Targeted panel sequencing of pharmacogenes and oncodrivers in colorectal cancer patients reveals genes with prognostic significance. Hum Genomics 2024; 18:83. [PMID: 39030589 PMCID: PMC11264515 DOI: 10.1186/s40246-024-00644-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 06/26/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Colorectal cancer is still the second leading cause of cancer-related deaths and thus biomarkers allowing prediction of the resistance of patients to therapy and estimating their prognosis are needed. We designed a panel of 558 genes with pharmacogenomics records related to 5-fluorouracil resistance, genes important for sensitivity to other frequently used drugs, major oncodrivers, and actionable genes. We performed a target enrichment sequencing of DNA from tumors and matched blood samples of patients, and compared the results with patient prognosis stratified by systemic adjuvant chemotherapy. RESULTS The median number of detected variants per tumor sample was 18.5 with 4 classified as having a high predicted functional effect and 14.5 moderate effect. APC, TP53, and KRAS were the most frequent mutated genes (64%, 59%, and 42% of mutated samples, respectively) followed by FAT4 (23%), FBXW7, and PIK3CA (16% for both). Patients with advanced stage III had more frequently APC, TP53, or KRAS mutations than those in stages I or II. KRAS mutation counts followed an increasing trend with grade (G1 < G2 < G3). The response to adjuvant therapy was worse in carriers of frameshift mutations in APC or 12D variant in KRAS, but none of these oncodrivers had prognostic value. Carriage of somatic mutations in any of the genes ABCA13, ANK2, COL7A1, NAV3, or UNC80 had prognostic relevance for worse overall survival (OS) of all patients. In contrast, mutations in FLG, GLI3, or UNC80 were prognostic in the same direction for patients untreated, and mutations in COL6A3, LRP1B, NAV3, RYR1, RYR3, TCHH, or TENM4 for patients treated with adjuvant therapy. The first association was externally validated. From all germline variants with high or moderate predicted functional effects (median 326 per patient), > 5% frequency and positive Manhattan plot based on 3-year RFS, rs72753407 in NFACS, rs34621071 in ERBB4, and rs2444274 in RIF1 were significantly associated with RFS, OS or both. CONCLUSIONS The present study identified several putative somatic and germline genetic events with prognostic potential for colorectal cancer that should undergo functional characterization.
Collapse
Affiliation(s)
- Lucie Heczko
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic
| | - Václav Liška
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Vyčítal
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic
- Department of Surgery, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Ondřej Fiala
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic
- Department of Oncology and Radiotherapeutics, Faculty of Medicine and University Hospital in Pilsen, Charles University, Pilsen, Czech Republic
| | - Simona Šůsová
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Viktor Hlaváč
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic.
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic.
| | - Pavel Souček
- Biomedical Center, Faculty of Medicine in Pilsen, Charles University, alej Svobody 1655/76, Pilsen, 323 00, Czech Republic.
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic.
| |
Collapse
|
16
|
Maurya R, Chug I, Vudatha V, Palma AM. Applications of spatial transcriptomics and artificial intelligence to develop integrated management of pancreatic cancer. Adv Cancer Res 2024; 163:107-136. [PMID: 39271261 DOI: 10.1016/bs.acr.2024.06.007] [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: 09/15/2024]
Abstract
Cancer is a complex disease intrinsically associated with cellular processes and gene expression. With the development of techniques such as single-cell sequencing and sequential fluorescence in situ hybridization (seqFISH), it was possible to map the location of cells based on their gene expression with more precision. Moreover, in recent years, many tools have been developed to analyze these extensive datasets by integrating machine learning and artificial intelligence in a comprehensive manner. Since these tools analyze sequencing data, they offer the chance to analyze any tissue regardless of its origin. By applying this to cancer settings, spatial transcriptomic analysis based on artificial intelligence may help us understand cell-cell communications within the tumor microenvironment. Another advantage of this analysis is the identification of new biomarkers and therapeutic targets. The integration of such analysis with other omics data and with routine exams such as magnetic resonance imaging can help physicians with the earlier diagnosis of tumors as well as establish a more personalized treatment for pancreatic cancer patients. In this review, we give an overview description of pancreatic cancer, describe how spatial transcriptomics and artificial intelligence have been used to study pancreatic cancer and provide examples of how integrating these tools may help physicians manage pancreatic cancer in a more personalized approach.
Collapse
Affiliation(s)
- Rishabh Maurya
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Isha Chug
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - António M Palma
- Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA, United States; VCU Institute of Molecular Medicine, Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
17
|
TAHIYA EC, ISLAM AA, HATTA M, LUSIKOOY RE, PRIHANTONO P, RUDIMAN R, WIDIANA IK, PATELONGI I, BUKHARI AS. 5-Fluorouracil for colorectal cancer: mechanism of action and metabolism. GAZZETTA MEDICA ITALIANA ARCHIVIO PER LE SCIENZE MEDICHE 2024; 183. [DOI: 10.23736/s0393-3660.23.05249-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
18
|
Abedpoor N, Taghian F, Jalali Dehkordi K, Safavi K. Sparassis latifolia and exercise training as complementary medicine mitigated the 5-fluorouracil potent side effects in mice with colorectal cancer: bioinformatics approaches, novel monitoring pathological metrics, screening signatures, and innovative management tactic. Cancer Cell Int 2024; 24:141. [PMID: 38637796 PMCID: PMC11027426 DOI: 10.1186/s12935-024-03328-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/12/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Prompt identification and assessment of the disease are essential for reducing the death rate associated with colorectal cancer (COL). Identifying specific causal or sensitive components, such as coding RNA (cRNA) and non-coding RNAs (ncRNAs), may greatly aid in the early detection of colorectal cancer. METHODS For this purpose, we gave natural chemicals obtained from Sparassis latifolia (SLPs) either alone or in conjunction with chemotherapy (5-Fluorouracil to a mouse colorectal tumor model induced by AOM-DSS. The transcription profile of non-coding RNAs (ncRNAs) and their target hub genes was evaluated using qPCR Real-Time, and ELISA techniques. RESULTS MSX2, MMP7, ITIH4, and COL1A2 were identified as factors in inflammation and oxidative stress, leading to the development of COL. The hub genes listed, upstream regulatory factors such as lncRNA PVT1, NEAT1, KCNQ1OT1, SNHG16, and miR-132-3p have been discovered as biomarkers for prognosis and diagnosis of COL. The SLPs and exercise, effectively decreased the size and quantity of tumors. CONCLUSIONS This effect may be attributed to the modulation of gene expression levels, including MSX2, MMP7, ITIH4, COL1A2, PVT1, NEAT1, KCNQ1OT1, SNHG16, and miR-132-3p. Ultimately, SLPs and exercise have the capacity to be regarded as complementing and enhancing chemotherapy treatments, owing to their efficacious components.
Collapse
Affiliation(s)
- Navid Abedpoor
- Department of Sports Physiology, Faculty of Sports Sciences, School of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Farzaneh Taghian
- Department of Sports Physiology, Faculty of Sports Sciences, School of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran.
| | - Khosro Jalali Dehkordi
- Department of Sports Physiology, Faculty of Sports Sciences, School of Sports Sciences, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| | - Kamran Safavi
- Department of Plant Biotechnology, Medicinal Plants Research Centre, Isfahan (Khorasgan) Branch, Islamic Azad University, Isfahan, Iran
| |
Collapse
|
19
|
Catozzi S, Assaad S, Delrieu L, Favier B, Dumas E, Hamy AS, Latouche A, Crochet H, Blay JY, Mullaert J, Ballesta A, Heudel P. Early morning immune checkpoint blockade and overall survival of patients with metastatic cancer: An In-depth chronotherapeutic study. Eur J Cancer 2024; 199:113571. [PMID: 38301362 DOI: 10.1016/j.ejca.2024.113571] [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: 09/21/2023] [Accepted: 01/15/2024] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Recent retrospective studies suggest potential large patient's benefit through proper timing of immune checkpoint blockers (ICB). The association between ICB treatment timing and patient survival, neoplastic response and toxicities was investigated, together with interactions with performance status (PS) and sex. METHODS A cohort of patients with metastatic or locally advanced solid tumors, who received pembrolizumab, nivolumab, atezolizumab, durvalumab, or avelumab, alone or with concomitant chemotherapy, between November 2015 and March 2021, at the Centre Leon Bérard (France), was retrospectively studied. RESULTS 361 patients were investigated (80% non-small cell lung cancer patients, mean [SD] age: 63 [11] years, 39% of women, 83% PS0-1 at first infusion, 19% received concomitant chemotherapy). ICB were administered from 07:25 to 17:21 and optimal morning/afternoon cut-off was 11:37. Morning infusions were associated with increased OS as compared to afternoon (median 30.3 vs 15.9 months, p = 0.0024; HR 1.56 [1.17-2.1], p = 0.003). A strong PS-timing interaction was found (PS0-1 patients, HR=1.53 [1.10-2.12], p = 0.011; PS2-3 patients, HR=0.50 [0.25-0.97], p = 0.042). Morning PS0-1 patients displayed increased OS (median 36.7 vs 21.3 months, p = 0.023), partial/complete response rate (58% vs 41%, p = 0.027), and grade1-3 toxicities (49% vs 34%, p = 0.028). Mortality risk ratio between infusions at worst time-of-day, estimated at 13:36 [12:48-14:23], and in early morning was equal to 4.8 ([2.3-10.1], p = 0.008). Timing differences in toxicities resulted significant only in female patients (women vs men: p < 0.001 vs 0.4). CONCLUSIONS Early morning ICB infusion was associated with increased OS, response, and toxicities in patients with PS0-1 as compared to later infusions within the day. Prospective randomized trials are needed to confirm this retrospective study.
Collapse
Affiliation(s)
- Simona Catozzi
- Inserm U900, Cancer Systems Pharmacology, Institut Curie, MINES ParisTech, CBIO, PSL Research University, 35 rue Dailly, 92250 Saint-Cloud, France
| | - Souad Assaad
- Département de cancérologie médicale, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Lidia Delrieu
- Residual Tumour and Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, Paris University, 75005 Paris, France
| | - Bertrand Favier
- Département de pharmacie oncologique, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Elise Dumas
- Residual Tumour and Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, Paris University, 75005 Paris, France
| | - Anne-Sophie Hamy
- Residual Tumour and Response to Treatment Laboratory, RT2Lab, Translational Research Department, INSERM, U932 Immunity and Cancer, Institut Curie, Paris University, 75005 Paris, France; Medical oncology, Université Paris, Institut Curie, Paris, France
| | - Aurélien Latouche
- INSERM U900, Statistical Methods for Precision Medicine Institut Curie, PSL Research University, 35 rue Dailly, Saint-Cloud, France; Conservatoire National des Arts et Métiers, Paris, France
| | - Hugo Crochet
- Direction des systèmes d'information, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| | - Jean-Yves Blay
- Département de cancérologie médicale, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France; Directeur général du Centre Léon Bérard, Université de Lyon, 28 rue Laennec, 69008 Lyon, France
| | - Jimmy Mullaert
- Faculty of Medicine, University of Versailles Saint-Quentin, Université Paris Saclay, 78000 Versailles, France. INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, 35 rue Dailly, 92210 Saint-Cloud, France
| | - Annabelle Ballesta
- Inserm U900, Cancer Systems Pharmacology, Institut Curie, MINES ParisTech, CBIO, PSL Research University, 35 rue Dailly, 92250 Saint-Cloud, France.
| | - Pierre Heudel
- Département de cancérologie médicale, Centre Léon Bérard, 28 rue Laennec, 69008 Lyon, France
| |
Collapse
|
20
|
Alalikhan A, Ebrahimi S, Aliee A, Mirzavi F, Hashemy SI. The combined anti-tumor effects of 5-fluorouracil and neurokinin receptor inhibitor, aprepitant, against colorectal cancer: In vitro and in vivo study. Med Oncol 2024; 41:70. [PMID: 38340190 DOI: 10.1007/s12032-024-02312-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the world's largest health concerns with growing global incidence and mortality. The potential value of the neurokinin-1 receptor as a therapeutic target has been reported in several tumor types, including CRC. Here we examined the potential anti-tumor effects of a clinically approved neurokinin-1 receptor antagonist, aprepitant, alone and its combination with 5-Fluorouracil (5-FU) as a first choice CRC chemotherapeutic drug, in both in vitro and in vivo models of CRC. METHODS MTT assay was employed for assessing cell proliferation. mRNA expression levels were determined by quantitative real-time PCR (qRT-PCR). Flow cytometric analysis of apoptosis was performed using an Annexin-V/propidium iodide assay kit. We finally conducted an in vivo experiment in a mouse model of CRC to confirm the in vitro antiproliferative activity of aprepitant and 5-FU. RESULTS We found that aprepitant and 5-FU significantly reduced CRC cell viability. The combination of drugs exhibited potent synergistic growth inhibitory effects on CRC cells. Moreover, aprepitant and 5-FU induced apoptosis and altered the levels of apoptotic genes (up-regulation of Bax, and p53 along with downregulation of Bcl-2). Importantly, the aprepitant and 5-FU combination showed a more pronounced impact on apoptosis and associated genes than either of the agents alone. Furthermore, aprepitant reduced tumor growth in vivo and led to significantly longer survival time, and this effect was more prominent when using the aprepitant and 5-FU combination. CONCLUSIONS Collectively, combinatory treatment with aprepitant and 5-FU potentially exerts synergistic growth inhibition and apoptosis induction in CRC, deserving further consideration as a novel strategy for CRC patients.
Collapse
Affiliation(s)
- Abbas Alalikhan
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safieh Ebrahimi
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Aliee
- Clinical Research Development Center, Imam Khomeini and Mohammad Kermanshahi and Farabi Hospitals, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
21
|
Lévi FA, Okyar A, Hadadi E, Innominato PF, Ballesta A. Circadian Regulation of Drug Responses: Toward Sex-Specific and Personalized Chronotherapy. Annu Rev Pharmacol Toxicol 2024; 64:89-114. [PMID: 37722720 DOI: 10.1146/annurev-pharmtox-051920-095416] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
Today's challenge for precision medicine involves the integration of the impact of molecular clocks on drug pharmacokinetics, toxicity, and efficacy toward personalized chronotherapy. Meaningful improvements of tolerability and/or efficacy of medications through proper administration timing have been confirmed over the past decade for immunotherapy and chemotherapy against cancer, as well as for commonly used pharmacological agents in cardiovascular, metabolic, inflammatory, and neurological conditions. Experimental and human studies have recently revealed sexually dimorphic circadian drug responses. Dedicated randomized clinical trials should now aim to issue personalized circadian timing recommendations for daily medical practice, integrating innovative technologies for remote longitudinal monitoring of circadian metrics, statistical prediction of molecular clock function from single-timepoint biopsies, and multiscale biorhythmic mathematical modelling. Importantly, chronofit patients with a robust circadian function, who would benefit most from personalized chronotherapy, need to be identified. Conversely, nonchronofit patients could benefit from the emerging pharmacological class of chronobiotics targeting the circadian clock.
Collapse
Affiliation(s)
- Francis A Lévi
- Chronotherapy, Cancers and Transplantation Research Unit, Faculty of Medicine, Paris-Saclay University, Villejuif, France;
- Gastrointestinal and General Oncology Service, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, France
- Department of Statistics, University of Warwick, Coventry, United Kingdom
| | - Alper Okyar
- Faculty of Pharmacy, Department of Pharmacology, Istanbul University, Beyazit-Istanbul, Turkey
| | - Eva Hadadi
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
- Laboratory for Myeloid Cell Immunology, Center for Inflammation Research VIB, Zwijnaarde, Belgium
| | - Pasquale F Innominato
- Oncology Department, Ysbyty Gwynedd Hospital, Betsi Cadwaladr University Health Board, Bangor, United Kingdom
- Warwick Medical School and Cancer Research Centre, University of Warwick, Coventry, United Kingdom
| | - Annabelle Ballesta
- Inserm Unit 900, Cancer Systems Pharmacology, Institut Curie, MINES ParisTech CBIO-Centre for Computational Biology, PSL Research University, Saint-Cloud, France
| |
Collapse
|
22
|
Amniouel S, Jafri MS. High-accuracy prediction of colorectal cancer chemotherapy efficacy using machine learning applied to gene expression data. Front Physiol 2024; 14:1272206. [PMID: 38304289 PMCID: PMC10830836 DOI: 10.3389/fphys.2023.1272206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/26/2023] [Indexed: 02/03/2024] Open
Abstract
Introduction: FOLFOX and FOLFIRI chemotherapy are considered standard first-line treatment options for colorectal cancer (CRC). However, the criteria for selecting the appropriate treatments have not been thoroughly analyzed. Methods: A newly developed machine learning model was applied on several gene expression data from the public repository GEO database to identify molecular signatures predictive of efficacy of 5-FU based combination chemotherapy (FOLFOX and FOLFIRI) in patients with CRC. The model was trained using 5-fold cross validation and multiple feature selection methods including LASSO and VarSelRF methods. Random Forest and support vector machine classifiers were applied to evaluate the performance of the models. Results and Discussion: For the CRC GEO dataset samples from patients who received either FOLFOX or FOLFIRI, validation and test sets were >90% correctly classified (accuracy), with specificity and sensitivity ranging between 85%-95%. In the datasets used from the GEO database, 28.6% of patients who failed the treatment therapy they received are predicted to benefit from the alternative treatment. Analysis of the gene signature suggests the mechanistic difference between colorectal cancers that respond and those that do not respond to FOLFOX and FOLFIRI. Application of this machine learning approach could lead to improvements in treatment outcomes for patients with CRC and other cancers after additional appropriate clinical validation.
Collapse
Affiliation(s)
- Soukaina Amniouel
- School of Systems Biology, George Mason University, Fairfax, VA, United States
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA, United States
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
23
|
Cayún JP, Cerpa LC, Colombo A, Cáceres DD, Leal JL, Reyes F, Gutiérrez-Cáceres C, Calfunao S, Varela NM, Quiñones LA. Genetic Polymorphisms and Tumoral Mutational Profiles over Survival in Advanced Colorectal Cancer Patients: An Exploratory Study. Curr Oncol 2024; 31:274-295. [PMID: 38248103 PMCID: PMC10814806 DOI: 10.3390/curroncol31010018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
Colorectal cancer is a common disease, both in Chile and worldwide. The most widely used chemotherapy schemes are based on 5-fluorouracil (5FU) as the foundational drug (FOLFOX, CapeOX). Genetic polymorphisms have emerged as potential predictive biomarkers of response to chemotherapy, but conclusive evidence is lacking. This study aimed to investigate the role of genetic variants associated with 5FU-based chemotherapy on therapeutic response, considering their interaction with oncogene mutations (KRAS, NRAS, PI3KCA, AKT1, BRAF). In a retrospective cohort of 63 patients diagnosed with metastatic colorectal cancer, a multivariate analysis revealed that liver metastases, DPYD, ABCB1, and MTHFR polymorphisms are independent indicators of poor prognosis, irrespective of oncogene mutations. BRAF wild-type status and high-risk drug-metabolism polymorphisms correlated with a poor prognosis in this Chilean cohort. Additionally, findings from the genomics of drug sensitivity (GDSC) project demonstrated that cell lines with wild-type BRAF have higher IC50 values for 5-FU compared to BRAF-mutated cell lines. In conclusion, the genetic polymorphisms DPYDrs1801265, ABCB1rs1045642, and MTHFRrs180113 may serve as useful biomarkers for predicting a poor prognosis in patients undergoing 5-fluorouracil chemotherapy, regardless of oncogene mutations.
Collapse
Affiliation(s)
- Juan Pablo Cayún
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago 8350499, Chile
| | - Leslie Carol Cerpa
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago 8350499, Chile
| | - Alicia Colombo
- Anatomy Pathology Service, Hospital Clínico de la Universidad de Chile, Santiago 8350499, Chile;
- Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile
| | - Dante Daniel Cáceres
- Institute of Population Health, School of Public Health, Faculty of Medicine, University of Chile, Santiago 8350499, Chile;
| | - José Luis Leal
- Cancer Research Department, Instituto Oncológico Fundación Arturo López Pérez, Santiago 8350499, Chile; (J.L.L.); (F.R.)
| | - Felipe Reyes
- Cancer Research Department, Instituto Oncológico Fundación Arturo López Pérez, Santiago 8350499, Chile; (J.L.L.); (F.R.)
| | - Carolina Gutiérrez-Cáceres
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Department of Pharmaceutical Sciences and Technology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8350499, Chile
| | - Susan Calfunao
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago 8350499, Chile
- Laboratory Pathological Anatomy, Hospital Luis Calvo Mackenna, Santiago 8350499, Chile
| | - Nelson Miguel Varela
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago 8350499, Chile
| | - Luis Abel Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic-Clinical Oncology (DOBC), Faculty of Medicine, University of Chile, Santiago 8350499, Chile; (J.P.C.); (L.C.C.); (C.G.-C.); (S.C.)
- Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago 8350499, Chile
- Department of Pharmaceutical Sciences and Technology, Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago 8350499, Chile
| |
Collapse
|
24
|
Bagheri M, Zandieh MA, Daryab M, Samaei SS, Gholami S, Rahmanian P, Dezfulian S, Eary M, Rezaee A, Rajabi R, Khorrami R, Salimimoghadam S, Hu P, Rashidi M, Ardakan AK, Ertas YN, Hushmandi K. Nanostructures for site-specific delivery of oxaliplatin cancer therapy: Versatile nanoplatforms in synergistic cancer therapy. Transl Oncol 2024; 39:101838. [PMID: 38016356 DOI: 10.1016/j.tranon.2023.101838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
As a clinically approved treatment strategy, chemotherapy-mediated tumor suppression has been compromised, and in spite of introducing various kinds of anticancer drugs, cancer eradication with chemotherapy is still impossible. Chemotherapy drugs have been beneficial in improving the prognosis of cancer patients, but after resistance emerged, their potential disappeared. Oxaliplatin (OXA) efficacy in tumor suppression has been compromised by resistance. Due to the dysregulation of pathways and mechanisms in OXA resistance, it is suggested to develop novel strategies for overcoming drug resistance. The targeted delivery of OXA by nanostructures is described here. The targeted delivery of OXA in cancer can be mediated by polymeric, metal, lipid and carbon nanostructures. The advantageous of these nanocarriers is that they enhance the accumulation of OXA in tumor and promote its cytotoxicity. Moreover, (nano)platforms mediate the co-delivery of OXA with drugs and genes in synergistic cancer therapy, overcoming OXA resistance and improving insights in cancer patient treatment in the future. Moreover, smart nanostructures, including pH-, redox-, light-, and thermo-sensitive nanostructures, have been designed for OXA delivery and cancer therapy. The application of nanoparticle-mediated phototherapy can increase OXA's potential in cancer suppression. All of these subjects and their clinical implications are discussed in the current review.
Collapse
Affiliation(s)
- Mohsen Bagheri
- Radiology Resident, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Babol Branch, Islamic Azad University, Babol, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Eary
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
25
|
Silva VR, Santos LDS, de Castro MVL, Dias RB, Valverde LDF, Rocha CAG, Soares MBP, Quadros CA, Correa RS, Batista AA, Bezerra DP. A novel ruthenium complex with 5-fluorouracil suppresses colorectal cancer stem cells by inhibiting Akt/mTOR signaling. Cell Death Discov 2023; 9:460. [PMID: 38104089 PMCID: PMC10725484 DOI: 10.1038/s41420-023-01759-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 11/20/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023] Open
Abstract
[Ru(5-FU)(PPh3)2(bipy)]PF6 (Ru/5-FU) is a novel ruthenium complex with 5-fluorouracil with promising potential against colorectal cancer (CRC). In the present study, we investigated the molecular mechanism of Ru/5-FU action in HCT116 CRC cells. Ru/5-FU exhibited potent cytotoxicity on a panel of cancer cell lines and on primary cancer cells and induced apoptosis in HCT116 CRC cells. Ru/5-FU reduced AKT1 gene transcripts, as well as the expression of Akt1 and Akt (pS473) and downstream Akt proteins mTOR (pS2448), S6 (pS235/pS236), 4EBP1 (pT36/pT45), GSK-3β (pS9) and NF-κB p65 (pS529), but not Akt upstream proteins Hsp90 and PI3K p85/p55 (pT458/pT199), indicating an inhibitory action of Akt/mTOR signaling. Ru/5-FU increased LC3B expression and reduced p62/SQSTM1 levels, indicating autophagy induction. Curiously, the autophagy inhibitors 3-methyladenine and chloroquine increased Ru/5-FU-induced cell death, indicating an induction of cytoprotective autophagy by this compound. Ru/5-FU also reduced clonogenic survival, as well as the percentage of CD133+ cells and colonosphere formation, indicating that Ru/5-FU can suppress stem cells in HCT116 cells. Ru/5-FU inhibited cell migration and invasion in wound healing assays and Transwell cell invasion assays, along with a reduction in vimentin expression and an increase in E-cadherin levels, indicating that Ru/5-FU can interfere with epithelial-mesenchymal transition. Ru/5-FU also inhibited in vivo HCT116 cell development and experimental lung metastases in mouse xenograft models. Altogether, these results indicate that Ru/5-FU is an anti-CRC chemotherapy drug candidate with the ability to suppress stemness in CRC cells by inhibiting Akt/mTOR signaling.
Collapse
Affiliation(s)
- Valdenizia R Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Luciano de S Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Maria V L de Castro
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Rosane B Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
| | - Ludmila de F Valverde
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
| | - Clarissa A G Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- Department of Propedeutics, School of Dentistry of the Federal University of Bahia, Salvador, Bahia, 40110-909, Brazil
| | - Milena B P Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil
- SENAI Institute of Innovation (ISI) in Health Advanced Systems, University Center SENAI/CIMATEC, Salvador, Bahia, 41650-010, Brazil
| | - Claudio A Quadros
- São Rafael Hospital, Rede D'Or/São Luiz, Salvador, Bahia, 41253-190, Brazil
- Bahia State University, Salvador, Bahia, 41150-000, Brazil
| | - Rodrigo S Correa
- Department of Chemistry, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, 35400-000, Brazil
| | - Alzir A Batista
- Department of Chemistry, Federal University of São Carlos, São Carlos, São Paulo, 13561-901, Brazil
| | - Daniel P Bezerra
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador, Bahia, 40296-710, Brazil.
| |
Collapse
|
26
|
Ding W, Wang JX, Wu JZ, Liu AC, Jiang LL, Zhang HC, Meng Y, Liu BY, Peng GJ, Lou EZ, Mao Q, Zhou H, Tang DL, Chen X, Liu JB, Shi XP. Targeting proteasomal deubiquitinases USP14 and UCHL5 with b-AP15 reduces 5-fluorouracil resistance in colorectal cancer cells. Acta Pharmacol Sin 2023; 44:2537-2548. [PMID: 37528233 PMCID: PMC10692219 DOI: 10.1038/s41401-023-01136-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 07/09/2023] [Indexed: 08/03/2023]
Abstract
5-Fluorouracil (5-FU) is the first-line treatment for colorectal cancer (CRC) patients, but the development of acquired resistance to 5-FU remains a big challenge. Deubiquitinases play a key role in the protein degradation pathway, which is involved in cancer development and chemotherapy resistance. In this study, we investigated the effects of targeted inhibition of the proteasomal deubiquitinases USP14 and UCHL5 on the development of CRC and resistance to 5-FU. By analyzing GEO datasets, we found that the mRNA expression levels of USP14 and UCHL5 in CRC tissues were significantly increased, and negatively correlated with the survival of CRC patients. Knockdown of both USP14 and UCHL5 led to increased 5-FU sensitivity in 5-FU-resistant CRC cell lines (RKO-R and HCT-15R), whereas overexpression of USP14 and UCHL5 in 5-FU-sensitive CRC cells decreased 5-FU sensitivity. B-AP15, a specific inhibitor of USP14 and UCHL5, (1-5 μM) dose-dependently inhibited the viability of RKO, RKO-R, HCT-15, and HCT-15R cells. Furthermore, treatment with b-AP15 reduced the malignant phenotype of CRC cells including cell proliferation and migration, and induced cell death in both 5-FU-sensitive and 5-FU-resistant CRC cells by impairing proteasome function and increasing reactive oxygen species (ROS) production. In addition, b-AP15 inhibited the activation of NF-κB pathway, suppressing cell proliferation. In 5-FU-sensitive and 5-FU-resistant CRC xenografts nude mice, administration of b-AP15 (8 mg·kg-1·d-1, intraperitoneal injection) effectively suppressed the growth of both types of tumors. These results demonstrate that USP14 and UCHL5 play an important role in the development of CRC and resistance to 5-FU. Targeting USP14 and UCHL5 with b-AP15 may represent a promising therapeutic strategy for the treatment of CRC.
Collapse
Affiliation(s)
- Wa Ding
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Jin-Xiang Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Precision Medicine Center, Department of Biobank, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jun-Zheng Wu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Ao-Chu Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Li-Ling Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Hai-Chuan Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Yi Meng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Bing-Yuan Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Guan-Jie Peng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - En-Zhe Lou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Qiong Mao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Huan Zhou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China
| | - Dao-Lin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xin Chen
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| | - Jin-Bao Liu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| | - Xian-Ping Shi
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Qingyuan, 511500, China.
| |
Collapse
|
27
|
Lei Y, Wang D, Chen W, Tian X, Wei J. FOXM1/NCAPH activates glycolysis to promote colon adenocarcinoma stemness and 5-FU resistance. Anticancer Drugs 2023; 34:929-938. [PMID: 37260271 DOI: 10.1097/cad.0000000000001526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Chemotherapy using 5-fluorouracil (5-FU) is currently considered the most effective treatment for advanced colon adenocarcinoma (COAD). However, drug resistance remains a major obstacle in treating COAD. Non-SMC condensin I complex subunit H ( NCAPH ) is known to have a certain impact on the development of COAD, but its precise involvement in the mechanism of 5-FU resistance has not been demonstrated. Bioinformatics analysis was utilized to assay the expression of NCAPH and Forkhead box M1 ( FOXM1 ) in COAD tumor tissues, which was then verified in COAD cell lines. The resistance of COAD cells to 5-FU was measured by CCK-8 assay, stemness was tested by cell sphere formation assay, and glycolysis ability was measured by cellular energy analysis metabolism. Chromatin Immunoprecipitation and dual-luciferase reporter assays were done to confirm the specific interaction between FOXM1 and NCAPH . The expression levels of FOXM1 and NCAPH were significantly upregulated in COAD tissues and cells, and they were involved in regulating the glycolytic signaling pathway. Inhibition of the glycolytic pathway could reverse the effect of NCAPH overexpression on COAD stemness and resistance. FOXM1 was identified as a transcription factor of NCAPH , and it regulated COAD glycolysis, cell stemness, and 5-FU resistance by activating NCAPH expression. FOXM1-mediated upregulation of NCAPH expression promoted COAD cell stemness and resistance via the glycolytic pathway. This study provides a possible mechanism for the FOXM1/NCAPH axis in the glycolytic pathway, cell stemness, and resistance in COAD.
Collapse
Affiliation(s)
- Yuehua Lei
- Department of General Surgery, Zigong Fourth People's Hospital, Zigong City, Sichuan Province, China
| | | | | | | | | |
Collapse
|
28
|
Hassan MM, Romana B, Mao G, Kumar N, Sonvico F, Thordarson P, Joyce P, Bremmell KE, Barnes TJ, Prestidge CA. Liposome-Micelle-Hybrid (LMH) Carriers for Controlled Co-Delivery of 5-FU and Paclitaxel as Chemotherapeutics. Pharmaceutics 2023; 15:1886. [PMID: 37514072 PMCID: PMC10385268 DOI: 10.3390/pharmaceutics15071886] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Paclitaxel (PTX) and 5-fluorouracil (5-FU) are clinically relevant chemotherapeutics, but both suffer a range of biopharmaceutical challenges (e.g., either low solubility or permeability and limited controlled release from nanocarriers), which reduces their effectiveness in new medicines. Anticancer drugs have several major limitations, which include non-specificity, wide biological distribution, a short half-life, and systemic toxicity. Here, we investigate the potential of liposome-micelle-hybrid (LMH) carriers (i.e., drug-loaded micelles encapsulated within drug-loaded liposomes) to enhance the co-formulation and delivery of PTX and 5-FU, facilitating new delivery opportunities with enhanced chemotherapeutic performance. We focus on the combination of liposomes and micelles for co-delivery of PTX and 5_FU to investigate increased drug loading, improved solubility, and transport/permeability to enhance chemotherapeutic potential. Furthermore, combination chemotherapy (i.e., containing two or more drugs in a single formulation) may offer improved pharmacological performance. Compared with individual liposome and micelle formulations, the optimized PTX-5FU-LMH carriers demonstrated increased drug loading and solubility, temperature-sensitive release, enhanced permeability in a Caco-2 cell monolayer model, and cancer cell eradication. LMH has significant potential for cancer drug delivery and as a next-generation chemotherapeutic.
Collapse
Affiliation(s)
- Md Musfizur Hassan
- School of Chemistry, The Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia
- School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Bilquis Romana
- School of Chemistry, The Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Guangzhao Mao
- School of Chemical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Naresh Kumar
- School of Chemistry, The Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Fabio Sonvico
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Pall Thordarson
- School of Chemistry, The Australian Centre for Nanomedicine, The University of New South Wales, Sydney, NSW 2052, Australia
| | - Paul Joyce
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Kristen E Bremmell
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Timothy J Barnes
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Clive A Prestidge
- Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| |
Collapse
|
29
|
Mehrzadi S, Sheibani M, Koosha F, Alinaghian N, Pourhanifeh MH, Tabaeian SAP, Reiter RJ, Hosseinzadeh A. Protective and therapeutic potential of melatonin against intestinal diseases: updated review of current data based on molecular mechanisms. Expert Rev Gastroenterol Hepatol 2023; 17:1011-1029. [PMID: 37796746 DOI: 10.1080/17474124.2023.2267439] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 10/03/2023] [Indexed: 10/07/2023]
Abstract
INTRODUCTION Intestinal diseases, a leading global cause of mortality and morbidity, carry a substantial socioeconomic burden. Small and large intestines play pivotal roles in gastrointestinal physiology and food digestion. Pathological conditions, such as gut dysbiosis, inflammation, cancer, therapy-related complications, ulcers, and ischemia, necessitate the urgent exploration of safe and effective complementary therapeutic strategies for optimal intestinal health. AREAS COVERED This article evaluates the potential therapeutic effects of melatonin, a molecule with a wide range of physiological actions, on intestinal diseases including inflammatory bowel disease, irritable bowel syndrome, colon cancer, gastric/duodenal ulcers and other intestinal disorders. EXPERT OPINION Due to anti-inflammatory and antioxidant properties as well as various biological actions, melatonin could be a therapeutic option for improving digestive disorders. However, more researches are needed to fully understand the potential benefits and risks of using melatonin for digestive disorders.
Collapse
Affiliation(s)
- Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Koosha
- Department of Radiology Technology, Faculty of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nazila Alinaghian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Serradilla-Martín M, Oliver-Guillén JR, Ruíz-Quijano P, Palomares-Cano A, de la Plaza-Llamas R, Ramia JM. Surgery of Colorectal Liver Metastases Involving the Inferior Vena Cava: A Systematic Review. Cancers (Basel) 2023; 15:2965. [PMID: 37296926 PMCID: PMC10251857 DOI: 10.3390/cancers15112965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Combined hepatic and inferior vena cava (IVC) resection is the only potentially curative treatment for patients with colorectal liver metastases (CRLM) involving the IVC. Most of the existing data come from case reports or small case series. In this paper, a systematic review based on the PICO strategy was performed in accordance with the PRISMA statement. Papers from January 1980 to December 2022 were searched in Embase, PubMed, and the Cochrane Library databases. Articles considered for inclusion had to present data on simultaneous liver and IVC resection for CRLM and report surgical and/or oncological outcomes. From a total of 1175 articles retrieved, 29, including a total of 188 patients, met the inclusion criteria. The mean age was 58.3 ± 10.8 years. The most frequent techniques used were right hepatectomy ± caudate lobe for hepatic resections (37.8%), lateral clamping (44.8%) for vascular control, and primary closure (56.8%) for IVC repair. The thirty-day mortality reached 4.6%. Tumour relapse was reported in 65.8% of the cases. The median overall survival (OS) was 34 months (with a confidence interval of 30-40 months), and the 1-year, 3-year, and 5-year OS were 71.4%, 19.8%, and 7.1%, respectively. In the absence of prospective randomized studies, which are difficult to perform, IVC resection seems to be safe and feasible.
Collapse
Affiliation(s)
- Mario Serradilla-Martín
- Department of Surgery, Instituto de Investigación Sanitaria Aragón, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | | | | | - Ana Palomares-Cano
- Department of Surgery, Hospital Universitario Miguel Servet, 50009 Zaragoza, Spain;
| | | | - José Manuel Ramia
- Department of Surgery, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain;
| |
Collapse
|
31
|
Chen JK, Merrick KA, Kong YW, Izrael-Tomasevic A, Eng G, Handly ED, Patterson JC, Cannell IG, Suarez-Lopez L, Hosios AM, Dinh A, Kirkpatrick DS, Yu K, Rose CM, Hernandez JM, Hwangbo H, Palmer AC, Vander Heiden MG, Yilmaz ÖH, Yaffe MB. An RNA Damage Response Network Mediates the Lethality of 5-FU in Clinically Relevant Tumor Types. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.28.538590. [PMID: 37162991 PMCID: PMC10168374 DOI: 10.1101/2023.04.28.538590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
5-fluorouracil (5-FU) is a successful and broadly used anti-cancer therapeutic. A major mechanism of action of 5-FU is thought to be through thymidylate synthase (TYMS) inhibition resulting in dTTP depletion and activation of the DNA damage response. This suggests that 5-FU should synergize with other DNA damaging agents. However, we found that combinations of 5-FU and oxaliplatin or irinotecan failed to display any evidence of synergy in clinical trials, and resulted in sub-additive killing in a panel of colorectal cancer (CRC) cell lines. In seeking to understand this antagonism, we unexpectedly found that an RNA damage response during ribosome biogenesis dominates the drug's efficacy in tumor types for which 5-FU shows clinical benefit. 5-FU has an inherent bias for RNA incorporation, and blocking this greatly reduced drug-induced lethality, indicating that accumulation of damaged RNA is more deleterious than the lack of new RNA synthesis. Using 5-FU metabolites that specifically incorporate into either RNA or DNA revealed that CRC cell lines and patient-derived colorectal cancer organoids are inherently more sensitive to RNA damage. This difference held true in cell lines from other tissues in which 5-FU has shown clinical utility, whereas cell lines from tumor tissues that lack clinical 5-FU responsiveness typically showed greater sensitivity to the drug's DNA damage effects. Analysis of changes in the phosphoproteome and ubiquitinome shows RNA damage triggers the selective ubiquitination of multiple ribosomal proteins leading to autophagy-dependent rRNA catabolism and proteasome-dependent degradation of ubiquitinated ribosome proteins. Further, RNA damage response to 5-FU is selectively enhanced by compounds that promote ribosome biogenesis, such as KDM2A inhibitors. These results demonstrate the presence of a strong RNA damage response linked to apoptotic cell death, with clear utility of combinatorially targeting this response in cancer therapy.
Collapse
Affiliation(s)
- Jung-Kuei Chen
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Karl A. Merrick
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yi Wen Kong
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - George Eng
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Erika D. Handly
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jesse C. Patterson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ian G. Cannell
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Lucia Suarez-Lopez
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aaron M. Hosios
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anh Dinh
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Kebing Yu
- Genentech Biotechnology company, South San Francisco, CA 94080, USA
| | | | - Jonathan M. Hernandez
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haeun Hwangbo
- Curriculum in Bioinformatics and Computational Biology, UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, Computational Medicine Program, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam C. Palmer
- Department of Pharmacology, Computational Medicine Program, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Matthew G. Vander Heiden
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Ömer H. Yilmaz
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Michael B. Yaffe
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Surgery, Beth Israel Medical Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
32
|
El-Hazek RMM, Zaher NH, Emam HES, El-Gazzar MG, Khalil A. Pyrazole-sulfonamide scaffold featuring dual-tail strategy as apoptosis inducers in colon cancer. Sci Rep 2023; 13:5782. [PMID: 37031294 PMCID: PMC10082777 DOI: 10.1038/s41598-023-32820-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/03/2023] [Indexed: 04/10/2023] Open
Abstract
Dual-tail strategy has been successfully utilized in the development of novel carbonic anhydrase IX (CA IX) inhibitors. Herein we adopted this approach in the design and synthesis of a series of novel pyridine sulfonamide-pyrazole hybrid scaffold mimicking dual-tail inhibitors of CA IX. A library of 15 compounds was synthesized and assessed for their potential cytotoxic effects against colorectal cancer cells. Compounds 3, and 11 induced potential cytotoxic effects against the three cancer cell lines (HCT-116, HT-29, and SW-620) with IC50s' of 45.88, 28.27, and 16.57 uM, 25.01, 8.99, and 3.27 µM, respectively. Both compounds induced cellular apoptosis on HCT-116 and SW-620 cells, while compound 3 induced necrosis as well. In addition, both compounds induced cell cycle arrest on G0/G1, and S phases. Also, compound 11 showed potential autophagy induction on both colon cancer cell lines (HCT-116, and HT-29), and a little bit on metastatic type. Both compounds were less cytotoxic than the reference drug on normal epithelial cell. The migration rates of HCT-116 and the metastatic one SW-620 were reduced by both compounds. Finally, molecular docking of compounds 3 and 11 into the active site of CA IX confirmed in vitro inhibitory activity for both compounds.
Collapse
Affiliation(s)
- Reham M M El-Hazek
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Nashwa H Zaher
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt
| | - Hagar E S Emam
- Biomedical Research Division, Nawah Scientific, Cairo, Egypt
| | - Marwa G El-Gazzar
- Drug Radiation Research Department, National Center for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority (EAEA), Cairo, 11787, Egypt.
| | - Amira Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt (BUE), El-Sherouk City, Cairo, 11837, Egypt.
| |
Collapse
|
33
|
Zhu J, Li G, Zhang Z, Wang Y. A Case of Complete Remission in Proficient Mismatch Repair (pMMR) Advanced Colon Cancer Treated with Sintilimab and XELOX. Immunotargets Ther 2023; 12:17-23. [PMID: 36844460 PMCID: PMC9951411 DOI: 10.2147/itt.s393526] [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: 10/27/2022] [Accepted: 02/09/2023] [Indexed: 02/20/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is the 3rd most common malignant tumors after breast cancer and lung cancer, accounting for 9.4% of patients. Some patients had distant metastasis at the time of diagnosis without surgery opportunity. It is particularly important to prolong patient survival and improve quality of life. Patient Concerns A 73-year-old female was admitted with discomfort over 2 months. Enlarged lymph nodes in the left supraclavicular fossa were observed in chest computed tomography (CT). Enhanced abdominal CT showed thickening of the right colon wall with multiple metastatic lymph nodes in the abdomen. Colonoscopy showed ileocecal mass and pathology showed moderately and poorly differentiated adenocarcinoma. Physical examination showed a 2*2 cm lymph node could be touched in the left supraclavicular fossa. The patient was diagnosed advanced colon cancer by the histopathological examination and imaging findings. Actually, it is hardly to resect radically. Intervention Sintilimab combined with XELOX was initiated. Two period of treatment after initial therapy, laparoscopic radical resection of right colon cancer was performed successfully. Outcomes After conversion treatment, the enlarged lymph nodes and primary tumor were significantly reduced. The patient was discharged successfully three weeks after surgery. Both specimen and 14 lymph nodes dissected showed no malignancy in pathology. Tumor regression grading (TRG) is 0, which indicate complete regression with no residual tumor cells including lymph nodes. The patient obtained a pathological complete response (pCR). Lessons The patient achieved a great therapeutic benefit with the above-mentioned chemotherapy in this case. The case provides a potential reference for pMMR CRC patients treating with immune checkpoint inhibitors (ICIs).
Collapse
Affiliation(s)
- Jiangpeng Zhu
- The Second People’s Hospital of Wuhu, Department of Gastrointestinal Surgery, East China Normal University, Wuhu, 241000, People’s Republic of China
| | - Guangyao Li
- The Second People’s Hospital of Wuhu, Department of Gastrointestinal Surgery, East China Normal University, Wuhu, 241000, People’s Republic of China,Correspondence: Guangyao Li, Department of Gastrointestinal Surgery, The Second People’s Hospital of Wuhu, East China Normal University, Wuhu, 241000, People’s Republic of China, Email
| | - Zhengjun Zhang
- The Second People’s Hospital of Wuhu, Department of Gastrointestinal Surgery, East China Normal University, Wuhu, 241000, People’s Republic of China
| | - Yandong Wang
- The Second People’s Hospital of Wuhu, Department of Gastrointestinal Surgery, East China Normal University, Wuhu, 241000, People’s Republic of China
| |
Collapse
|
34
|
Xanthones from Gentianella acuta (Michx.) Hulten Ameliorate Colorectal Carcinoma via the PI3K/Akt/mTOR Signaling Pathway. Int J Mol Sci 2023; 24:ijms24032279. [PMID: 36768602 PMCID: PMC9917368 DOI: 10.3390/ijms24032279] [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: 01/06/2023] [Revised: 01/17/2023] [Accepted: 01/21/2023] [Indexed: 01/26/2023] Open
Abstract
Colorectal carcinoma (CRC) is a kind of malignant tumor closely related to ulcerative colitis. Xanthone derivatives are one of the most promising therapeutic drugs which have been used in phase I/II clinical trials for cancer therapy. Our previous study indicated that the aerial parts of Gentianella acuta Michx. Hulten (GA) was rich in xanthones and showed a good therapeutic effect on ulcerative colitis in mice, suggesting that GA xanthones might have some therapeutic or ameliorative effects on CRC. However, no relevant study has been reported. This study aims to find the effective substances of GA inhibiting CRC and clarify their mechanism. Solvent extraction, column chromatographic separation, and LC-MS analysis were used to characterize the 70% EtOH extract of GA and track xanthones abundant fraction XF. MTT assay was carried out to clarify the activity of GA fractions; the result showed XF to be the main active fraction. LC-MS analysis was executed to characterize XF, 38 xanthones were identified. Network pharmacology prediction, in vitro activity screening, and molecular docking assay were combined to predict the potential mechanism; the PI3K/Akt/mTOR signaling pathway was found to be most important. Western blot assay on the main active xanthones 1,3,5-trihydroxyxanthone (16), 1,3,5,8-tetrahydroxyxanthone (17), 1,5,8-trihydroxy-3-methoxyxanthone (18), and 1,7-dihydroxy-3,8-dimethoxyxanthone (19) was used to verify the above prediction; these xanthones were found to inhibit the PI3K/Akt/mTOR signaling pathway, and 17 played a significant role among them through Western blot assay using PI3K/AKT/mTOR agonist IGF-1. In conclusion, this study demonstrated that GA xanthones were effective compounds of GA inhibiting CRC by regulating PI3K/Akt/mTOR signaling pathway transduction, at least. Importantly, 1,3,5,8-tetrahydroxyxanthone (17), the most abundant active xanthone in GA, might be a candidate drug for CRC.
Collapse
|
35
|
Szota M, Wolski P, Carucci C, Marincola FC, Gurgul J, Panczyk T, Salis A, Jachimska B. Effect of Ionization Degree of Poly(amidoamine) Dendrimer and 5-Fluorouracil on the Efficiency of Complex Formation-A Theoretical and Experimental Approach. Int J Mol Sci 2023; 24:ijms24010819. [PMID: 36614260 PMCID: PMC9821593 DOI: 10.3390/ijms24010819] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/05/2023] Open
Abstract
Due to their unique structure, poly(amidoamine) (PAMAM) dendrimers can bind active ingredients in two ways: inside the structure or on their surface. The location of drug molecules significantly impacts the kinetics of active substance release and the mechanism of internalization into the cell. This study focuses on the effect of the protonation degree of the G4PAMAM dendrimer and the anticancer drug 5-fluorouracil (5FU) on the efficiency of complex formation. The most favorable conditions for constructing the G4PAMAM-5FU complex are a low degree of protonation of the dendrimer molecule with the drug simultaneously present in a deprotonated form. The fluorine components in the XPS spectra confirm the formation of the stable complex. Through SAXS and DLS methods, a decrease in the dendrimer's molecular size resulting from protonation changes at alkaline conditions was demonstrated. The gradual closure of the dendrimer structure observed at high pH values makes it difficult for the 5FU molecules to migrate to the interior of the support structure, thereby promoting drug immobilization on the surface. The 1H NMR and DOSY spectra indicate that electrostatic interactions determine the complex formation process. Through MD simulations, the localization profile and the number of 5FU molecules forming the complex were visualized on an atomic scale.
Collapse
Affiliation(s)
- Magdalena Szota
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Krakow, Poland
| | - Pawel Wolski
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Krakow, Poland
| | - Cristina Carucci
- Department of Chemical and Geological Sciences, University of Cagliari, 09042 Cagliari, Italy
| | | | - Jacek Gurgul
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Krakow, Poland
| | - Tomasz Panczyk
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Krakow, Poland
| | - Andrea Salis
- Department of Chemical and Geological Sciences, University of Cagliari, 09042 Cagliari, Italy
| | - Barbara Jachimska
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, 30-239 Krakow, Poland
- Correspondence:
| |
Collapse
|
36
|
Villette CC, Orrell D, Millen J, Chassagnole C. Should personalised dosing have a role in cancer treatment? Front Oncol 2023; 13:1154493. [PMID: 37213297 PMCID: PMC10196464 DOI: 10.3389/fonc.2023.1154493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/13/2023] [Indexed: 05/23/2023] Open
Abstract
Almost all pharmaceutical products are approved on the basis of their effect in patients representing the "average" of the population studied in registrational trials, with most drug labels allowing, at most, for empirical dose reduction in the case of toxicity. In this perspective article we explore some of the evidence that supports the use of personalised dosing in cancer treatment and show how we have been able to build on existing models linking dose, exposure and toxicity to demonstrate how dose optimisation, including increasing the dose, has the potential to significantly improve efficacy outcomes. We also explore, through the lens of our own experience of developing a personalised dosing platform, some of the hurdles that stand in the way of implementing a personalised approach to dosing in real world settings. In particular, our experience is illustrated by the application of a dosing platform for docetaxel treatment in prostate cancer.
Collapse
|
37
|
Kdimati S, Bürtin F, Linnebacher M, Mullins CS. Patient-Derived Organoids for In Vivo Validation of In Vitro Data. Methods Mol Biol 2023; 2589:111-126. [PMID: 36255621 DOI: 10.1007/978-1-0716-2788-4_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Patient-derived organoids are promising tumor models for functional validation of next-generation sequencing-based therapy recommendations. In times of rapidly advancing precision oncology approaches in everyday clinical processes, reliable and valid tumor models are required. Tumor organoids consist of tumor "stem" cells, differentiated (epithelial) tumor, and stroma cells. The cellular architecture and interactions closely mimic the original patient tumor. These organoids can be implanted into immunodeficient mice, generating patient-derived organoid-derived xenografts, thus enabling in vitro to in vivo transfer. Most importantly, the high clinical relevance of PDO models is maintained in this conversion. This protocol describes in detail the methods and techniques as well as the materials necessary to generate in vitro PDO and in vivo PDO-derived xenograft models. The elaborate process description starts with the processing of freshly obtained tumor tissue. The proceedings include tissue processing, organoid culturing, PDO implantation into immunodeficient mice, tumor explantation, and finally tumor preservation. All these proceedings are described in this timely chronological order. This protocol will enable researchers to generate PDO models from freshly received tumor tissue and generate PDO-derived xenografts. Models generated according to these methods are suitable for a very broad research spectrum.
Collapse
Affiliation(s)
- Said Kdimati
- Department of General Surgery, Molecular Oncology and Immunotherapy, Rostock, Germany
| | - Florian Bürtin
- Department of General Surgery, Molecular Oncology and Immunotherapy, Rostock, Germany
| | - Michael Linnebacher
- Department of General Surgery, Molecular Oncology and Immunotherapy, Rostock, Germany
| | | |
Collapse
|
38
|
Identifying the Role of Oxidative Stress-Related Genes as Prognostic Biomarkers and Predicting the Response of Immunotherapy and Chemotherapy in Ovarian Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6575534. [PMID: 36561981 PMCID: PMC9764017 DOI: 10.1155/2022/6575534] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 10/14/2022] [Indexed: 12/14/2022]
Abstract
Background Ovarian cancer (OC) is one of the most frequently seen and fatal gynecological malignancies, and oxidative stress (OS) plays a critical role in the development and chemoresistance of OC. Materials and Methods OS-related genes (OSRGs) were obtained from the Molecular Signatures Database. Besides, gene expression profiles and clinical information from The Cancer Genome Atlas (TCGA) were selected to identify the prognostic OSRGs. Moreover, univariate Cox regression, LASSO, and multivariate Cox regression analyses were conducted sequentially to establish a prognostic signature, which was later validated in three independent Gene Expression Omnibus (GEO) datasets. Next, gene set enrichment analysis (GSEA) and tumor mutation burden (TMB) analysis were performed. Afterwards, immune checkpoint genes (ICGs) and the tumor immune dysfunction and exclusion (TIDE) algorithm, together with IMvigor210 and GSE78220 cohorts, were applied to comprehensively explore the role of OSRG signature in immunotherapy. Further, the CellMiner and Genomics of Drug Sensitivity in Cancer (GDSC) databases were also applied in investigating the significance of OSRG signature in chemotherapy. Results Altogether, 34 prognostic OSRGs were identified, among which 14 were chosen to establish the most valuable prognostic signature. The Kaplan-Meier (KM) analysis suggested that patients with lower OS-related risk score had better prognosis. The area under the curve (AUC) values were 0.71, 0.76, and 0.85 in 3, 5, and 7 years separately, and the stability of this prognostic signature was confirmed in three GEO datasets. As revealed by GSEA and TMB analysis results, OC patients in low-risk group might have better immunotherapeutic response, which was consistent with ICG expression and TIDE analyses. Moreover, both IMvigor210 and GSE78220 cohorts demonstrated that patients with lower OS-related risk score were more likely to benefit from anti-PD-1/L1 immunotherapy. In addition, the association between prognostic signature and drug sensitivity was explored. Conclusion According to our results in this work, OSRG signature can act as a powerful prognostic predictor for OC, which contributes to generating more individualized therapeutic strategies for OC patients.
Collapse
|
39
|
Teufel A, Li M, Gerken M, Ebert MP, Schlitt HJ, Evert M, Herr W, Klinkhammer-Schalke M. Second Cancer After Additive Chemotherapy in Patients With Colon Cancer. Clin Colorectal Cancer 2022; 21:354-361. [PMID: 35934636 DOI: 10.1016/j.clcc.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 12/18/2021] [Accepted: 07/03/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Additive chemotherapeutic treatment of UICC-stage -III / IV colon cancer with fluorouracil, leucovorin and oxaliplatin is widely accepted as current standard of treatment after R0-resection. However, as patients.. survival is increasing, long-term side effects of chemotherapeutic agents such as second cancer development are becoming increasingly important. PATIENTS We therefore investigated a total of 2 856 Patients with UICC-stage III / IV colon cancer, 223 of whom (7.8%) had developed a subsequent second cancer. RESULTS Median follow-up was 73.2 months (range 209.9 months, 95%-CI 69.8-76.9). Most frequent second cancers were prostate cancer (18.4%), colon cancer (16.1%), breast cancers (8.1%), lung cancer (8.1%), rectal cancer (4.9%) and uterine cancer (4.9%). However, in comparison to non-treated patients this did not represent a significantly increased risk for subsequent second cancer in patients after treatment with additive chemotherapy. Of interest, our data suggest a significantly decreased second cancer rate in patients treated with FOLFOX compared to FUFOL for additive treatment. CONCLUSIONS Second cancer development was not increased after additive chemotherapy for colon cancer, which is a novel aspect in the ongoing discussions on reduction of adjuvant treatment to 3 months or treatment of lymph node negative patients. Novelty and Impact Statement To our knowledge, this is the first population-based study analyzing second cancer development after additive chemotherapy in patients with UICC III-IV colon cancer. The results have an important impact on the surveillance and long-term follow-up of cancer patients.
Collapse
Affiliation(s)
- Andreas Teufel
- Department of Medicine II, Division of Hepatology, Division of Clinical Bioinformatics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Moying Li
- Department of Medicine II, Division of Hepatology, Division of Clinical Bioinformatics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Gerken
- Regensburg Tumor Center, Institute for Quality Assurance and Health Services Research at the University of Regensburg, Regensburg, Germany
| | - Matthias P Ebert
- Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPDBW), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Matthias Evert
- Department of Pathology, University of Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Medical Center Regensburg, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Regensburg Tumor Center, Institute for Quality Assurance and Health Services Research at the University of Regensburg, Regensburg, Germany
| |
Collapse
|
40
|
Al-Hujaily EM, Al-Sowayan BS, Alyousef Z, Uddin S, Alammari F. Recruiting Immunity for the Fight against Colorectal Cancer: Current Status and Challenges. Int J Mol Sci 2022; 23:13696. [PMID: 36430176 PMCID: PMC9697544 DOI: 10.3390/ijms232213696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/31/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Cancer immunotherapies have changed the landscape of cancer management and improved the standard treatment protocols used in multiple tumors. This has led to significant improvements in progression-free survival and overall survival rates. In this review article, we provide an insight into the major immunotherapeutic methods that are currently under investigation for colorectal cancer (CRC) and their clinical implementations. We emphasize therapies that are based on monoclonal antibodies (mAbs) and adoptive cell therapy, their mechanisms of action, their advantages, and their potential in combination therapy. We also highlight the clinical trials that have demonstrated both the therapeutic efficacy and the toxicities associated with each method. In addition, we summarize emerging targets that are now being evaluated as potential interventions for CRC. Finally, we discuss current challenges and future direction for the cancer immunotherapy field.
Collapse
Affiliation(s)
- Ensaf M. Al-Hujaily
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Batla S. Al-Sowayan
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Zeyad Alyousef
- Department of Surgery, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh 14611, Saudi Arabia
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Farah Alammari
- Department of Blood and Cancer Research, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Clinical Laboratory Sciences Department, College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| |
Collapse
|
41
|
Lin X, Xu L, Tan H, Zhang X, Shao H, Yao L, Huang X. The potential effects and mechanisms of Gegen Qinlian Decoction in oxaliplatin-resistant colorectal cancer based on network pharmacology. Heliyon 2022; 8:e11305. [DOI: 10.1016/j.heliyon.2022.e11305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022] Open
|
42
|
Sutton TL, Wong LH, Walker BS, Dewey EN, Eil R, Lopez CD, Kardosh A, Chen EY, Rocha FG, Billingsley KG, Mayo SC. Hepatectomy is associated with improved oncologic outcomes in recurrent colorectal liver metastases: A propensity-matched analysis. Surgery 2022; 173:1314-1321. [DOI: 10.1016/j.surg.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 11/24/2022]
|
43
|
Hassan ESE, Shafaa MW, Faraag AHI, Essawy E, Bakkar AA, Al-Megrin WA, El-Khadragy MF, Abdelfattah MS, Abdel Moneim AE. Evaluation of the antineoplastic property of prodigiosins and 5-fluorouracil in restraining the growth of Ehrlich solid tumors in mice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:83723-83732. [PMID: 35773616 DOI: 10.1007/s11356-022-21678-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 06/21/2022] [Indexed: 06/15/2023]
Abstract
Prodigiosins have been shown to have anticancer activities. 5-Fluorouracil (5-FU) is broadly used chemotherapeutic drug that treats different solid tumors including breast cancer but has low response rates and a variety of side effects. In this study, we evaluated the anticancer properties of prodigiosins in a murine model "Ehrlich tumor" and tested whether it can be added to 5-FU to potentiate its effects. Markers of oxidative stress; MDA, NO, and GSH levels were evaluated as well as antioxidant enzyme activities of CAT SOD, GR, and GPx. The levels of Bax, Bcl-2, PCNA, and NF-κB proteins were measured using ELISA kits. The mRNAs of p53 and Cdc2 and Casp3 were quantitatively measured by real-time PCR and ELISA respectively. Cell cycle analysis was performed using flow cytometery. Prodigiosins did not influence tumor volume. Prodigiosins have not induced oxidative stress while 5-FU did increase MDA, NO but decreased GSH levels. The combination prodigiosins and 5-FU did reduce oxidative stress markers; MDA, NO and increased GSH levels. Prodigiosins significantly increased CAT only while 5-FU did decreased SOD, CAT, GPx, and GR. The combination prodigiosins and 5-FU increased the levels of these enzymes again. Prodigiosins increased the Bax/Bcl-2 ratio while the combination deceased it. In conclusion, prodigiosins have pronounced anticancer properties but their combination with 5-FU decreased oxidative stress exerted by 5-FU but weakened the apoptotic effects of 5-FU. Prodigiosins could affect a key mechanism through which 5-FU exerts its tumor inhibitory effects.
Collapse
Affiliation(s)
- Elsayed S E Hassan
- Department of Chemistry, Faculty of Science, Helwan University, Cairo, 11795, Egypt
| | - Medhat W Shafaa
- Medical biophysics Division, Physics Department, Faculty of Science Helwan University, Cairo, 11795, Egypt
| | - Ahmed H I Faraag
- Botany and Microbiology Department, Faculty of Science, Helwan University, Cairo, 11795, Egypt
| | - Ehab Essawy
- Botany and Microbiology Department, Faculty of Science, Helwan University, Cairo, 11795, Egypt
| | - Ashraf A Bakkar
- Faculty of Biotechnology, Modern Sciences and Arts University (MSA), Giza, Egypt
| | - Wafa A Al-Megrin
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia
| | - Manal F El-Khadragy
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh, 11671, Saudi Arabia.
| | | | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo, 11795, Egypt
| |
Collapse
|
44
|
Ludikhuize MC, Gevers S, Nguyen NTB, Meerlo M, Roudbari SKS, Gulersonmez MC, Stigter ECA, Drost J, Clevers H, Burgering BMT, Rodríguez Colman MJ. Rewiring glucose metabolism improves 5-FU efficacy in p53-deficient/KRAS G12D glycolytic colorectal tumors. Commun Biol 2022; 5:1159. [PMID: 36316440 PMCID: PMC9622833 DOI: 10.1038/s42003-022-04055-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 09/30/2022] [Indexed: 11/29/2022] Open
Abstract
Despite the fact that 5-fluorouracil (5-FU) is the backbone for chemotherapy in colorectal cancer (CRC), the response rates in patients is limited to 50%. The mechanisms underlying 5-FU toxicity are debated, limiting the development of strategies to improve its efficacy. How fundamental aspects of cancer, such as driver mutations and phenotypic heterogeneity, relate to the 5-FU response remains obscure. This largely relies on the limited number of studies performed in pre-clinical models able to recapitulate the key features of CRC. Here, we analyzed the 5-FU response in patient-derived organoids that reproduce the different stages of CRC. We find that 5-FU induces pyrimidine imbalance, which leads to DNA damage and cell death in the actively proliferating cancer cells deficient in p53. Importantly, p53-deficiency leads to cell death due to impaired cell cycle arrest. Moreover, we find that targeting the Warburg effect in KRASG12D glycolytic tumor organoids enhances 5-FU toxicity by further altering the nucleotide pool and, importantly, without affecting non-transformed WT cells. Thus, p53 emerges as an important factor in determining the 5-FU response, and targeting cancer metabolism in combination with replication stress-inducing chemotherapies emerges as a promising strategy for CRC treatment. In p53-deficient colorectal cancer organoids, 5-fluorouracil induces pyrimidine imbalance, which causes DNA damage and cell death. Rewiring glucose metabolism through PDK inhibition by DCA enhances 5-FU toxicity in glycolytic p53-deficient organoids.
Collapse
Affiliation(s)
- Marlies C. Ludikhuize
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Sira Gevers
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Nguyen T. B. Nguyen
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Maaike Meerlo
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - S. Khadijeh Shafiei Roudbari
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - M. Can Gulersonmez
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Edwin C. A. Stigter
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Jarno Drost
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands ,grid.499559.dOncode Institute, Utrecht, The Netherlands
| | - Hans Clevers
- grid.487647.ePrincess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands ,grid.499559.dOncode Institute, Utrecht, The Netherlands ,grid.418101.d0000 0001 2153 6865Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, The Netherlands
| | - Boudewijn M. T. Burgering
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands ,grid.418101.d0000 0001 2153 6865Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, The Netherlands
| | - Maria J. Rodríguez Colman
- grid.7692.a0000000090126352Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| |
Collapse
|
45
|
A phase I/II study of arfolitixorin and 5-fluorouracil in combination with oxaliplatin (plus or minus bevacizumab) or irinotecan in metastatic colorectal cancer. ESMO Open 2022; 7:100589. [PMID: 36183444 PMCID: PMC9588906 DOI: 10.1016/j.esmoop.2022.100589] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/18/2022] [Accepted: 08/21/2022] [Indexed: 11/21/2022] Open
Abstract
Background 5-fluorouracil (5-FU) combined with a folate remains an essential treatment component for metastatic colorectal cancer (mCRC). Leucovorin is the folate most often used, but requires intracellular conversion to a reduced folate, and has high pharmacokinetic variability and limited bioavailability in patients with low folate pathway gene expression. Arfolitixorin is an immediately active form of folate, [6R]-5,10-methylenetetrahydrofolate ([6R]-MTHF), and may improve outcomes. Patients and methods This open-label, multicenter, phase I/II study in patients with mCRC (NCT02244632) assessed the tolerability and efficacy of first- or second-line arfolitixorin (30, 60, 120, or 240 mg/m2 intravenous) with 5-FU alone, or in combination with oxaliplatin (plus or minus bevacizumab) or irinotecan, every 14 days. Safety, efficacy, and pharmacokinetics were assessed before and after four cycles (8 weeks) of treatment. Results In 105 treated patients, investigators reported 583 adverse events (AEs) in 86 patients (81.9%), and 256 AEs (43.9%) were potentially related to arfolitixorin and 5-FU. Dose adjustments were required in 16 patients (15.2%). At 8 weeks, 9 out of 57 patients assessed for efficacy achieved an objective response (15.8%), and all 9 achieved a partial response. Six of these nine patients had received arfolitixorin as a first-line treatment. A further 33 patients (57.9%) achieved stable disease. Pharmacokinetics were assessed in 35 patients. The average tmax was 10 min, and area under the plasma concentration–time curve from time 0 to 1 h increased linearly between 30 and 240 mg/m2. No accumulation was observed for [6R]-MTHF following repeated administration, and there were no major pharmacokinetic differences between cycle 1 and cycle 4 at any dose. Conclusions Arfolitixorin is a well-tolerated moderator of 5-FU activity. It is suitable for further investigation in mCRC and has the potential to improve treatment outcomes in patients with low folate pathway gene expression. Arfolitixorin can easily be incorporated into current standard of care, requiring minimal changes to chemotherapy regimens.
Arfolitixorin is an immediately active form of folate and may improve outcomes with 5-FU-based chemotherapy. This phase I/II clinical trial demonstrated that arfolitixorin is a well-tolerated and effective folate agent in mCRC. Arfolitixorin can easily be incorporated into the current standard of care and is suitable for further investigation.
Collapse
|
46
|
Buyana B, Naki T, Alven S, Aderibigbe BA. Nanoparticles Loaded with Platinum Drugs for Colorectal Cancer Therapy. Int J Mol Sci 2022; 23:11261. [PMID: 36232561 PMCID: PMC9569963 DOI: 10.3390/ijms231911261] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is a common cancer in both men and women. Numerous studies on the therapeutic effectiveness of nanoparticles against colorectal cancer have been reported. Platinum treatments as well as other medications comprising of nanoparticles have been utilized. Drug resistance restricts the use of platinum medicines, despite their considerable efficacy against a variety of cancers. This review reports clinically licensed platinum medicines (cisplatin, carboplatin, and oxaliplatin) combined with various nanoparticles that have been evaluated for their therapeutic efficacy in the treatment of colorectal cancer, including their mechanism of action, resistance, and limitations.
Collapse
Affiliation(s)
| | | | | | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice 5700, Eastern Cape Province, South Africa
| |
Collapse
|
47
|
Castellani G, Buccarelli M, Lulli V, Ilari R, De Luca G, Pedini F, Boe A, Felli N, Biffoni M, Pilozzi E, Marziali G, Ricci-Vitiani L. MiR-378a-3p Acts as a Tumor Suppressor in Colorectal Cancer Stem-Like Cells and Affects the Expression of MALAT1 and NEAT1 lncRNAs. Front Oncol 2022; 12:867886. [PMID: 35814429 PMCID: PMC9263271 DOI: 10.3389/fonc.2022.867886] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
MiR-378a-3p plays a critical role in carcinogenesis acting as a tumor suppressor, promoting apoptosis and cell cycle arrest and reducing invasion and drug resistance in several human cancers, including colorectal cancer (CRC), where its expression is significantly associated with histological classification and prognosis. In this study, we investigated the biological and cellular processes affected by miR-378a-3p in the context of CRC carcinogenesis. In agreement with the literature, miR-378a-3p is downregulated in our cohort of CRC patients as well as, in 15 patient-derived colorectal cancer stem-like cell (CRC-SC) lines and 8 CRC cell lines, compared to normal mucosae. Restoration of miR-378a-3p restrains tumorigenic properties of CRC and CRC-SC lines, as well as, significantly reduces tumor growth in two CRC-SC xenograft mouse models. We reported that miR-378a-3p modulates the expression of the lncRNAs MALAT1 and NEAT1. Their expression is inversely correlated with that of miR-378a-3p in patient-derived CRC-SC lines. Silencing of miR-378a-3p targets, MALAT1 and NEAT1, significantly impairs tumorigenic properties of CRC-SCs, supporting the critical role of miR-378a-3p in CRC carcinogenesis as a tumor-suppressor factor by establishing a finely tuned crosstalk with lncRNAs MALAT1 and NEAT1.
Collapse
Affiliation(s)
- Giorgia Castellani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Valentina Lulli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Ramona Ilari
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gabriele De Luca
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pedini
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Core Facilities, Istituto Superiore di Sanità, Rome, Italy
| | - Nadia Felli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Mauro Biffoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, UOC Anatomia Patologica, Sant’Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Giovanna Marziali
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- *Correspondence: Lucia Ricci-Vitiani,
| |
Collapse
|
48
|
Huang X, Ke K, Jin W, Zhu Q, Zhu Q, Mei R, Zhang R, Yu S, Shou L, Sun X, Feng J, Duan T, Mou Y, Xie T, Wu Q, Sui X. Identification of Genes Related to 5-Fluorouracil Based Chemotherapy for Colorectal Cancer. Front Immunol 2022; 13:887048. [PMID: 35784334 PMCID: PMC9247273 DOI: 10.3389/fimmu.2022.887048] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/16/2022] [Indexed: 12/22/2022] Open
Abstract
Background Colorectal cancer (CRC) is one of the most common malignancies and its incidence and mortality are increasing yearly. 5-Fluorouracil (5-FU) has long been used as a standard first-line treatment for CRC patients. Although 5-FU-based chemotherapy is effective for advanced CRC, the consequent resistance remains a key problem and causes the poor prognosis of CRC patients. Thus, there is an urgent need to identify new biomarkers to predict the response to 5-FU-based chemotherapy. Methods CRC samples were retrieved from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA). The immune-related genes were retrieved from the ImmPort database. Single-cell sequencing results from colorectal cancer were obtained by the ArrayExpress database. 5-FU resistance-related genes were filtered and validated by R packages. ESTIMATE algorithms were used to assess the tumor microenvironment (TME). KEGG and GO analysis were performed to explore the biological signaling pathway for resistant-response patients and sensitive-response patients in the tumor microenvironment. pRRophetic algorithms were used to predict 5-FU sensitivity. GSEA and GSVA analysis was performed to excavate the biological signaling pathway of the RBP7 gene. Results Nine immune-related genes were identified to be associated with 5-FU resistance and poor disease-free survival (DFS) of CRC patients and the signature of these genes was developed in a DFS-prognostic model. Four immune-related genes were determined to be associated with 5-FU resistance and overall survival (OS) of CRC patients. The signature of these genes was developed an OS-prognostic model. ESTIMATE scores showed a significant difference between 5-FU resistant and 5-FU sensitive CRC patients. Resistant-response patients and sensitive-response patients to 5-FU based chemotherapy showed different GO and KEGG enrichment on the tumor microenvironment. RBP7, as a tumor immune microenvironment (TIME) related gene, was found to have the potential of predicting chemotherapy resistance and poor prognosis of CRC patients. GSEA analysis showed multiple signaling differences between the high and low expression of RBP7 in CRC patients. Hypoxia and TNFα signaling via NFκB gene sets were significantly different between chemotherapy resistant (RBP7High) and chemotherapy sensitive (RBP7Low) patients. Single-cell RNA-seq suggested RBP7 was centrally distributed in endothelial stalk cells, endothelial tip cells, and myeloid cells. Conclusions Immune-related genes will hopefully be potential prognostic biomarkers to predict chemotherapy resistance for CRC. RBP7 may function as a tumor microenvironment regulator to induce 5-FU resistance, thereby affecting the prognosis of CRC patients.
Collapse
Affiliation(s)
- Xingxing Huang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Kun Ke
- Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Weiwei Jin
- Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qianru Zhu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Qicong Zhu
- Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ruyi Mei
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Ruonan Zhang
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Shuxian Yu
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Lan Shou
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xueni Sun
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Jiao Feng
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Ting Duan
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yiping Mou
- Department of Gastrointestinal-Pancreatic Surgery, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Qibiao Wu, ; Xinbing Sui,
| | - Tian Xie
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Qibiao Wu, ; Xinbing Sui,
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Guangdong-Hong Kong-Macau Joint Laboratory for Contaminants Exposure and Health, Guangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Qibiao Wu, ; Xinbing Sui,
| | - Xinbing Sui
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
- School of Pharmacy and Department of Medical Oncology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
- *Correspondence: Yiping Mou, ; Tian Xie, ; Qibiao Wu, ; Xinbing Sui,
| |
Collapse
|
49
|
Talaat IM, Yakout NM, Soliman AS, Venkatachalam T, Vinod A, Eldohaji L, Nair V, Hareedy A, Kandil A, Abdel-Rahman WM, Hamoudi R, Saber-Ayad M. Evaluation of Galanin Expression in Colorectal Cancer: An Immunohistochemical and Transcriptomic Study. Front Oncol 2022; 12:877147. [PMID: 35707368 PMCID: PMC9190230 DOI: 10.3389/fonc.2022.877147] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/27/2022] [Indexed: 01/02/2023] Open
Abstract
Colorectal cancer (CRC) represents around 10% of all cancers, with an increasing incidence in the younger age group. The gut is considered a unique organ with its distinctive neuronal supply. The neuropeptide, human galanin, is widely distributed in the colon and expressed in many cancers, including the CRC. The current study aimed to explore the role of galanin at different stages of CRC. Eighty-one CRC cases (TNM stages I - IV) were recruited, and formalin-fixed paraffin-embedded samples were analyzed for the expression of galanin and galanin receptor 1 (GALR1) by immunohistochemistry (IHC). Galanin intensity was significantly lower in stage IV (n= 6) in comparison to other stages (p= 0.037 using the Mann-Whitney U test). Whole transcriptomics analysis using NGS was performed for selected samples based on the galanin expression by IHC [early (n=5) with high galanin expression and late (n=6) with low galanin expression]. Five differentially regulated pathways (using Absolute GSEA) were identified as drivers for tumor progression and associated with higher galanin expression, namely, cell cycle, cell division, autophagy, transcriptional regulation of TP53, and immune system process. The top shared genes among the upregulated pathways are AURKA, BIRC5, CCNA1, CCNA2, CDC25C, CDK2, CDK6, EREG, LIG3, PIN1, TGFB1, TPX2. The results were validated using real-time PCR carried out on four cell lines [two primaries (HCT116 and HT29) and two metastatic (LoVo and SK-Co-1)]. The current study shows galanin as a potential negative biomarker. Galanin downregulation is correlated with advanced CRC staging and linked to cell cycle and division, autophagy, transcriptional regulation of TP53 and immune system response.
Collapse
Affiliation(s)
- Iman M. Talaat
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nada M. Yakout
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | - Thenmozhi Venkatachalam
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Physiology and Immunology, College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Arya Vinod
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Leen Eldohaji
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Vidhya Nair
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Amal Hareedy
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Alaa Kandil
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Alexandria University, Cairo, Egypt
| | - Wael M. Abdel-Rahman
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Rifat Hamoudi
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Division of Surgery and Interventional Science, University College London, London, United Kingdom
| | - Maha Saber-Ayad
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Pharmacology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
50
|
Okuno K, Garg R, Yuan YC, Tokunaga M, Kinugasa Y, Goel A. Berberine and Oligomeric Proanthocyanidins Exhibit Synergistic Efficacy Through Regulation of PI3K-Akt Signaling Pathway in Colorectal Cancer. Front Oncol 2022; 12:855860. [PMID: 35600365 PMCID: PMC9114748 DOI: 10.3389/fonc.2022.855860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Naturally occurring dietary botanicals offer time-tested safety and anti-cancer efficacy, and a combination of certain compounds has shown to overcome the elusive chemotherapeutic resistance, which is of great significance for improving the mortality of patients with colorectal cancer (CRC). Accordingly, herein, we hypothesized that berberine (BBR) and oligomeric proanthocyanidins (OPCs) might regulate synergistically multiple oncogenic pathways to exert a superior anti-cancer activity in CRC. METHODS We performed a series of cell culture studies, followed by their interrogation in patient-derived organoids to evaluate the synergistic effect of BBR and OPCs against CRC. In addition, by performing whole genome transcriptomic profiling we identified the key targeted genes and pathways regulated by the combined treatment. RESULTS We first demonstrated that OPCs facilitated enhanced cellular uptake of BBR in CRC cells by measuring the fluorescent signal of BBR in cells treated individually or their combination. The synergism between BBR and OPCs were investigated in terms of their anti-tumorigenic effect on cell viability, clonogenicity, migration, and invasion. Furthermore, the combination treatment potentiated the cellular apoptosis in an Annexin V binding assay. Transcriptomic profiling identified oncogene MYB in PI3K-AKT signaling pathway might be critically involved in the anti-tumorigenic properties of the combined treatment. Finally, we successfully validated these findings in patient-derived CRC tumor organoids. CONCLUSIONS Collectively, we for the first time demonstrate that a combined treatment of BBR and OPCs synergistically promote the anti-tumorigenic properties in CRC possibly through the regulation of cellular apoptosis and oncogene MYB in the PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Keisuke Okuno
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, United States
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Rachana Garg
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, United States
| | - Yate-Ching Yuan
- Translational Bioinformatics, Center for Informatics, City of Hope, Duarte, CA, United States
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA, United States
- City of Hope Comprehensive Cancer Center, Duarte, CA, United States
| |
Collapse
|