1
|
Ultimo A, Jain A, Gomez-Gonzalez E, Alex TS, Moreno-Borrallo A, Jana S, Ghosh S, Ruiz-Hernandez E. Nanotherapeutic Formulations for the Delivery of Cancer Antiangiogenics. Mol Pharm 2025. [PMID: 40184281 DOI: 10.1021/acs.molpharmaceut.4c00822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2025]
Abstract
Antiangiogenic medications for cancer treatment have generally failed in showing substantial benefits in terms of prolonging life on their own; their effects are noticeable only when combined with chemotherapy. Moreover, treatments based on prolonged antiangiogenics administration have demonstrated to be ineffective in stopping tumor progression. In this scenario, nanotherapeutics can address certain issues linked to existing antiangiogenic treatments. More specifically, they can provide the ability to target the tumor's blood vessels to enhance drug accumulation and manage release, ultimately decreasing undesired side effects. Additionally, they enable the administration of multiple angiogenesis inhibitors at the same time as chemotherapy. Key reports in this field include the design of polymeric nanoparticles, inorganic nanoparticles, vesicles, and hydrogels for loading antiangiogenic substances like endostatin and interleukin-12. Furthermore, nanoformulations have been proposed to efficiently control relevant pro-angiogenic pathways such as VEGF, Tie2/Angiopoietin-1, HIF-1α/HIF-2α, and TGF-β, providing powerful approaches to block tumor growth and metastasis. In this article, we outline a selection of nanoformulations for antiangiogenic treatments for cancer that have been developed in the past ten years.
Collapse
Affiliation(s)
- Amelia Ultimo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Ayushi Jain
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Elisabet Gomez-Gonzalez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Thomson Santosh Alex
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Almudena Moreno-Borrallo
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Sukanya Jana
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| | - Shubhrima Ghosh
- Trinity Translational Medicine Institute, Trinity College Dublin, the University of Dublin, St. James's Hospital, Dublin 8 D08 NHY1, Ireland
- School of Biological, Health and Sports Sciences, Technological University Dublin, Grangegorman Lower, Dublin 7 D07 ADY7, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, the University of Dublin, College Green, Dublin 2 D02 PN40, Ireland
| |
Collapse
|
2
|
Zhou L, Huang X, Shi J, Yang Y, Dong F, Wei H, Ji C, Shan Y. Decoding colorectal cancer targeted therapy: a bibliometric journey of the last decade (2015-2024). Discov Oncol 2025; 16:442. [PMID: 40169453 PMCID: PMC11961832 DOI: 10.1007/s12672-025-02251-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
Colorectal cancer remains one of the most commonly diagnosed cancers globally, with a significant impact on public health. Targeted therapies have revolutionized the treatment landscape for colorectal cancer by offering increased specificity and reduced systemic toxicity compared to conventional chemotherapy. This study provides a comprehensive bibliometric analysis of global research on targeted therapy for colorectal cancer, focusing on publications from 2015 to 2024. A total of 3213 publications were retrieved from the Web of Science Core Collection and analyzed using bibliometric tools to construct knowledge maps and visualize research trends. The regression analysis shows a strong upward trend in publications from 2015 to 2024 (P < 0.001, R2 = 0.889). China leads in publication output, with the University of Texas MD Anderson Cancer Center contributing the highest number of studies. Tabernero and Kopetz are the core authors in the field. Research in this domain has primarily concentrated on the development and clinical assessment of drugs targeting the EGFR, RAS, VEGF, and BRAF signaling pathways, as well as investigating the pathogenesis, drug resistance, and metastatic mechanisms of colorectal cancer. Current advancements emphasize Artificial Intelligence-driven multi-omics integration, the creation of novel therapeutics targeting established molecular pathways, and the execution of global clinical trials to validate personalized treatment strategies.
Collapse
Affiliation(s)
- Linpo Zhou
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Xuanwei Huang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Jing Shi
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Yebin Yang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Fanhe Dong
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Haoran Wei
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Chenghao Ji
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, 310053, Zhejiang, China
| | - Yuqiang Shan
- Department of Gastrointestinal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
3
|
García-Alfonso P, Valladares-Ayerbes M, Muñoz Martín AJ, Morales Herrero R, Galvez Muñoz E, Prat-Llorens G. State of the art of the molecular hyperselection to guide treatment with anti-EGFR antibodies in RAS WT mCRC: implications for clinical practice and future perspectives. Expert Opin Biol Ther 2025; 25:413-423. [PMID: 40066702 DOI: 10.1080/14712598.2025.2477192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION Adding monoclonal antibodies to chemotherapy drastically changed the landscape of advanced colorectal cancer. The prediction of benefit from anti-EGFR therapies is mainly based on the absence of mutations in RAS and BRAF genes, the primary tumor sidedness and microsatellite MSS/MSI status. Molecular hyperselection may optimize the outcome of patients receiving anti-EGFR while detecting additional resistance alterations, both in chemo-naïve and in chemo-refractory settings. AREAS COVERED Our review focuses on negative molecular hyperselection, both on tissue samples and ctDNA, and the impact of this further patient selection on response rate and survival outcomes. We searched electronic database, selecting relevant English-language publications from 2017 to 2024. EXPERT OPINION Negative hyperselection beyond RAS and BRAF in advanced colorectal cancer appears to be a powerful tool for predicting outcomes to anti-EGFR therapy and spare patients from unnecessary treatment. This improvement appears in both naïve and pre-treated patients. However, data come mainly from retrospective studies. Therefore, to validate and integrate these findings in the clinical practice, prospective studies should be conducted. It will be interesting to elucidate the role of ctDNA in this setting and the choice of molecular techniques, considering costs and accessibility, to guarantee its implementation in the clinic.
Collapse
Affiliation(s)
- Pilar García-Alfonso
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Manuel Valladares-Ayerbes
- Medical Oncology Department, Hospital Universitario Virgen del Rocío/Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | - Andrés J Muñoz Martín
- Medical Oncology Service, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Rocío Morales Herrero
- Medical Oncology Department, Hospital Universitario Virgen del Rocío/Instituto de Biomedicina de Sevilla (IBIS), Sevilla, Spain
| | | | | |
Collapse
|
4
|
Zhang JY, Li H, Zhang MJ, Sun ZJ. Lymphangiogenesis orchestrating tumor microenvironment: Face changing in immunotherapy. Biochim Biophys Acta Rev Cancer 2025; 1880:189278. [PMID: 39929379 DOI: 10.1016/j.bbcan.2025.189278] [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: 11/13/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/19/2025]
Abstract
In the era of immunotherapy, the lymphatic system has garnered significant attention from researchers. Increasing evidence highlights the complex regulation of lymphatic vessels (LVs) within the tumor microenvironment, unveiling a paradox in tumor progression: while LVs enhance immune surveillance, they simultaneously foster immune suppression. This review summarizes the regulatory factors of lymphangiogenesis, discusses the intricate effects of LVs on immunotherapy, and emphasizes the potential connection between lymphangiogenesis and tertiary lymphoid structure. Additionally, current therapeutic strategies targeting lymphangiogenesis are critically evaluated, with a forward-looking perspective on future research directions and regulatory approaches to achieve precise targeting and optimize immunotherapy paradigms.
Collapse
Affiliation(s)
- Jun-Ye Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
| | - Hao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, China
| | - Meng-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan 430079, China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Centre for Immunology and Metabolism, Taikang Centre for Life and Medical Sciences, Wuhan University, Wuhan 430079, China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Wuhan 430079, China.
| |
Collapse
|
5
|
Sigler GI, Murtha J, Varley PR. Diagnostic Advances and Novel Therapeutics in Peritoneal Metastasis. Surg Oncol Clin N Am 2025; 34:173-194. [PMID: 40015798 DOI: 10.1016/j.soc.2024.12.005] [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: 03/01/2025]
Abstract
Appropriate assessment of disease burden in patients with peritoneal surface malignancy (PSM) is critical for treatment decision-making, and conventional cross-sectional imaging (computed tomography and/or MRI) often underestimates burden of disease. Advances in imaging for PSM include novel functional imaging modalities that target cells unique to the tumor microenvironment. Novel alternative methods of diagnosis and disease monitoring are also potentially applicable to management of PSM. These include forms of "liquid biopsy" targeting circulating tumor DNA. Novel regional therapies include both new therapeutic agents (immune-based and nanoparticle-based), as well as new methods of delivery such as pressurized intraperitoneal aerosolized chemotherapy.
Collapse
Affiliation(s)
- Gregory I Sigler
- Division of Surgical Oncology, Department of General Surgery, Complex General Surgical Oncology, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Jacqueline Murtha
- Department of General Surgery, General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA
| | - Patrick R Varley
- Division of Surgical Oncology, Department of General Surgery, University of Wisconsin School of Medicine and Public Health, 600 Highland Avenue, Mail Code 7375, Madison, WI 53792, USA; William S. Middleton Memorial Veterans Affairs Hospital, Madison, WI, USA.
| |
Collapse
|
6
|
Kato Y. Lenvatinib enhances antitumor immunity of anti-PD-1 antibody. Int J Clin Oncol 2025; 30:666-673. [PMID: 39985645 PMCID: PMC11946938 DOI: 10.1007/s10147-025-02721-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/04/2025] [Indexed: 02/24/2025]
Abstract
Lenvatinib is an orally available multi-tyrosine kinase inhibitor that mainly targets vascular endothelial growth factor (VEGF) and fibroblast growth factor (FGF) signaling. These inhibitory activities of lenvatinib exhibit antitumor efficacy, mainly due to their repressive effects on angiogenesis. In addition, a recent non-clinical evaluation using mouse tumor models revealed that lenvatinib causes immunomodulatory effects, including activation of effector T-cells and regulation of tumor-associated macrophages (TAMs). Combined treatment with lenvatinib and anti-programmed cell death-1 antibody (anti-PD-1) resulted in enhanced antitumor activity relative to monotreatment with anti-PD-1 or lenvatinib. This review summarizes the antitumor mechanisms of lenvatinib and of lenvatinib plus anti-PD-1 combination therapy.
Collapse
Affiliation(s)
- Yu Kato
- Tsukuba Research Laboratories, Eisai Co., Ltd., Tsukuba, Ibaraki, Japan.
| |
Collapse
|
7
|
Hsu CY, Altalbawy FMA, Oghenemaro EF, Uthirapathy S, Chandra M, Nathiya D, Kaur P, Ravi Kumar M, Kadhim AJ, Kariem M. Exosomal lncRNAs in the Tumor Angiogenesis: As Therapeutic Targets in Cancer Treatment. Arch Pharm (Weinheim) 2025; 358:e202400940. [PMID: 40165644 DOI: 10.1002/ardp.202400940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/12/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Exosomes, as mediators of intercellular communication, can be released from different types of cells and regulate the function of the target cell by transferring cargo, such as proteins, DNA, and RNA. Recent investigations have revealed a preponderance of long noncoding RNAs (lncRNAs), a subclass of noncoding RNAs, within exosomes, where they exhibit notable stability and are implicated in the development and progression of neoplastic processes, such as tumor angiogenesis. Angiogenesis, as a hallmark of cancer, provides diffusible nutrients and oxygen to the distant cells and guarantees tumorigenesis and metastasis. Exosomal lncRNAs, including MALAT1, OIP5-AS1, PART1, SNHG family, FAM225A, ATB, RAMP2-AS1, UCA1, TRPM2-AS, FGD5-AS1, and LINC0016, could modulate tumor angiogenesis by activating signaling cascades and mediators within the target cells, such as microRNAs (miRNAs). Regulation of tumor angiogenesis through modulation of exosomal lncRNAs could be a reliable strategy for cancer therapy. In this review, we discuss the characteristics and biogenesis of exosomes and lncRNAs and how exosomal lncRNAs are involved in various processes of tumorigenesis. Our primary focus is on exosomal lncRNAs, their impact on tumor angiogenesis, and their potential as novel diagnostic markers and therapeutic targets for various cancers.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, Egypt
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Muktesh Chandra
- Marwadi University Research Center, Department of Bioinformatics, Faculty of Engineering and Technology, Marwadi University, Rajkot, Gujarat, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
8
|
Huijbers EJM, van Beijnum JR, van Loon K, Griffioen CJ, Volckmann R, Bassez A, Lambrechts D, Nunes Monteiro M, Jimenez CR, Hogendoorn PCW, Koster J, Griffioen AW. Embryonic reprogramming of the tumor vasculature reveals targets for cancer therapy. Proc Natl Acad Sci U S A 2025; 122:e2424730122. [PMID: 40096611 PMCID: PMC11962416 DOI: 10.1073/pnas.2424730122] [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: 11/28/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
A sustained blood supply is critical for tumor growth, as it delivers the nutrients and oxygen required for development. Targeting of blood vessel formation via immunotherapies is an area of great importance. Knowing that certain embryonic genes, such as carcinoembryonic antigens (CEA) and oncofetal fibronectin, become reexpressed in malignant transformation, we hypothesized that a similar phenomenon holds true for tumor endothelial cells (TECs) as well. An approach for identification of highly selective tumor endothelial markers was conducted to develop targeted antiangiogenic immunotherapies. We first queried the transcriptome that is present during embryo development. We then performed a systematic search for genes selectively expressed in the mouse embryo at days E11 and E18, as compared to the transcriptome of the adult mouse. Subsequently, we queried for expression of these embryonic genes in sorted murine TECs. This approach identified among others the tumor endothelial antigens fibrillin-2 (Fbn2), elastin microfibril interface-located protein 2 (Emilin2) as well as the tumor endothelial antigens lysyl oxidase (Lox) and serine/cysteine protease inhibitor, clade E, member 1 (Serpine1; Pai-1). For these selected genes, functional involvement in angiogenesis was confirmed in in vitro bioassays. We subsequently used iBoost conjugate vaccine technology to develop vaccines against the selected targets. For all four targets, vaccination readily induced target-specific antibody responses in mice, resulting in inhibition of tumor growth. Access to highly specific tumor endothelial markers provides opportunities for direct targeting of the tumor vasculature with high specificity, without affecting healthy vasculature.
Collapse
Affiliation(s)
- Elisabeth J. M. Huijbers
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| | - Judy R. van Beijnum
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Karlijn van Loon
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Christian J. Griffioen
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Richard Volckmann
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Ayse Bassez
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Diether Lambrechts
- Vlaams Instituut voor Biotechnologie-Katolieke Universiteit Leuven Center for Cancer Biology, Leuven3000, Belgium
| | - Madalena Nunes Monteiro
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | - Connie R. Jimenez
- Department of Medical Oncology, Oncoproteomics Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
| | | | - Jan Koster
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1105 AZ, The Netherlands
| | - Arjan W. Griffioen
- Department of Medical Oncology, Angiogenesis Laboratory, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam1081 HV, The Netherlands
- CimCure Besloten Vennootschap, Amsterdam1066 CX, The Netherlands
| |
Collapse
|
9
|
Pakvisal N, Goldberg RM, Sathitruangsak C, Silaphong W, Faengmon S, Teeyapun N, Teerapakpinyo C, Tanasanvimon S. Overall survival with frontline vs subsequent anti-epidermal growth factor receptor therapies in unresectable, RAS/BRAF wild-type, left-sided metastatic colorectal cancer. World J Clin Oncol 2025; 16:102076. [PMID: 40130051 PMCID: PMC11866077 DOI: 10.5306/wjco.v16.i3.102076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/25/2024] [Accepted: 12/12/2024] [Indexed: 01/21/2025] Open
Abstract
BACKGROUND The combination of anti-epidermal growth factor receptor (EGFR) therapy and chemotherapy is currently a preferred first-line treatment for patients with unresectable, RAS and BRAF wild-type, left-sided metastatic colorectal cancer (mCRC). Several studies have also demonstrated the benefit of anti-EGFR therapy in subsequent line settings for this patient population. However, direct evidence comparing the effectiveness of frontline vs subsequent anti-EGFR therapy remains limited, leaving a crucial gap in guiding optimal treatment strategies. AIM To compare overall survival (OS) between frontline and subsequent anti-EGFR treatment in patients with unresectable, RAS and BRAF wild-type, left-sided mCRC. METHODS We retrospectively reviewed the medical records of mCRC patients treated at The King Chulalongkorn Memorial Hospital and Songklanagarind Hospital, Thailand, between January 2013 and April 2023. Patients were classified into two groups based on the sequence of their anti-EGFR treatment. The primary endpoint was OS. RESULTS Among 222 patients with a median follow-up of 29 months, no significant difference in OS was observed between the frontline and subsequent-line groups (HR 1.03, 95%CI: 0.73-1.46, P = 0.878). The median OS was 35.53 months (95%CI: 26.59-44.47) for the frontline group and 31.60 months (95%CI: 27.83-35.37) for the subsequent-line group. In the subsequent-line group, 71 patients (32.4%) who ultimately never received anti-EGFR therapy had a significantly worse median OS of 19.70 months (95%CI: 12.87-26.53). CONCLUSION Frontline and subsequent-line anti-EGFR treatments provide comparable OS in unresectable, RAS/BRAF wild-type, left-sided mCRC patients, but early exposure is vital for those unlikely to receive subsequent therapy.
Collapse
Affiliation(s)
- Nussara Pakvisal
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Richard M Goldberg
- Department of Medicine, WVU Cancer Institute, West Virginia University, Morgantown, WV 26506, United States
| | - Chirawadee Sathitruangsak
- Medical Oncology Unit, Division of Internal Medicine, Faculty of Medicine, Holistic Center for Cancer Study and Care (HOCC-PSU) and Prince of Songkla University, Songkhla 90110, Thailand
| | - Witthaya Silaphong
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Satawat Faengmon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Nattaya Teeyapun
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Chinachote Teerapakpinyo
- Chulalongkorn GenePRO Center, Research Affairs, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| | - Suebpong Tanasanvimon
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and The King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand
| |
Collapse
|
10
|
Masuda T, Funakoshi T, Horimatsu T, Masui S, Hira D, Inoue M, Yajima K, Nakagawa S, Ikemi Y, Hamanishi J, Takai A, Yamamoto S, Matsubara T, Mandai M, Seno H, Yanagita M, Muto M, Terada T, Yonezawa A. Population pharmacokinetic analysis of bevacizumab in Japanese cancer patients with proteinuria: a prospective cohort study. Cancer Chemother Pharmacol 2025; 95:46. [PMID: 40116970 DOI: 10.1007/s00280-025-04769-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/06/2025] [Indexed: 03/23/2025]
Abstract
PURPOSE Bevacizumab (BV) is an effective therapeutic antibody utilized in various cancers. Serum BV concentration can be a factor that potentially affects its therapeutic efficacy. Although proteinuria could affect BV pharmacokinetics, its influence was not evaluated in the previous population pharmacokinetic (PopPK) studies. Because BV can cause proteinuria as an adverse event, the present study aimed to develop a PopPK model in patients with proteinuria and to evaluate the influence of proteinuria on BV pharmacokinetics. METHODS This prospective cohort study enrolled 70 Japanese cancer patients newly starting BV, and 368 concentration samples from these patients were analyzed. Serum BV concentrations were measured at several time points including at the onset of proteinuria. PopPK analysis was conducted using a non-linear mixed-effects modeling program. A two-compartment model was used to estimate total body clearance (CL). RESULTS Serum BV concentrations divided by the dose per body weight and dosing interval tended to be lower in patients with higher urinary protein to creatinine ratio (UPCR). The covariate analysis showed that increasing BV CL was associated with decreasing serum albumin concentration and increasing body weight and UPCR. The simulated median trough concentrations of BV in patients with Common Terminology Criteria for Adverse Events grades 1, 2, and 3 proteinuria were decreased by 12.0%, 20.6%, and 31.5%, respectively, compared to those in patients with grade 0. CONCLUSION We successfully established a PopPK model incorporating UPCR to predict serum BV concentrations in patients with proteinuria. Our study provides additional insights to better understand BV pharmacokinetics.
Collapse
Affiliation(s)
- Takashi Masuda
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Taro Funakoshi
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takahiro Horimatsu
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sho Masui
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Daiki Hira
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Marin Inoue
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Kodai Yajima
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan
| | - Shunsaku Nakagawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Yasuaki Ikemi
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Takai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Yamamoto
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takeshi Matsubara
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoko Yanagita
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Terada
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan
| | - Atsushi Yonezawa
- Department of Clinical Pharmacology and Therapeutics, Kyoto University Hospital, Kyoto, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
- Division of Integrative Clinical Pharmacology, Faculty of Pharmacy, Keio University, 1-5-30 Shiba Koen, Minato-Ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
11
|
Zhao L, Zhang Z, Wang D, Yang L, Liu Z, Lou C. Analysis of the effectiveness and safety of lenvatinib/bevacizumab combined with PD-1/PD-L1 inhibitors and GEMOX in the first-line treatment of advanced biliary tract carcinoma. Clin Exp Med 2025; 25:87. [PMID: 40106138 PMCID: PMC11922973 DOI: 10.1007/s10238-025-01623-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
To assess the efficacy and safety of lenvatinib/bevacizumab combined with programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors and gemcitabine/oxaliplatin (GEMOX) as first-line treatments in patients with advanced biliary tract cancer (BTC). Patients with advanced BTC who received lenvatinib/bevacizumab combined with PD-1/PD-L1 inhibitors plus gemcitabine/oxaliplatin (GEMOX) chemotherapy were retrospectively screened. The primary endpoints were overall survival (OS) and progression-free survival (PFS), whereas the secondary endpoints were objective response rate (ORR), disease control rate (DCR), and safety. Prognostic factors for survival were analyzed. A total of 172 individuals were enrolled and categorized into four groups: Group A received GEMOX plus PD-1 antibody (sintilimab or camrelizumab) and lenvatinib; group B received GEMOX and PD-1 antibody (sintilimab or camrelizumab) and bevacizumab; group C received GEMOX and PD-1 antibody (sintilimab or camrelizumab); and group D received GEMOX alone. The median OS was 13.63 months (95% confidence interval [CI]: 12.37-14.89), 12.41 months (95% CI: 10.67-12.32), 11.23 months (95% CI: 9.39-13.07), and 8.86 months (95% CI: 7.28-10.44) in groups A, B, C, and D, respectively (P = 0.312). In groups A, B, C, and D, the median PFS was 12.42 months, 11.05 months, 8.89 months, and 6.02 months. A statistically significant difference was observed (t = 2, 95% CI: 11.31-13.53, P < 0.01). The ORR was 45.00% (17/40) in group A, 34.78% (16/46) in group B, 16.67% (5/30) in group C, and 17.86% (10/56) in group D. The DCR was 87.50% (35/40), 78.26% (36/46), 76.67% (23/30), and 58.93% (33/56) in groups A, B, C, and D, respectively. In addition, regression analysis showed that patients' metastasis site, whether the neutrophil-lymphocyte ratio was < 2.3, and whether chemotherapy was administered through hepatic artery embolization and was independent prognostic factors influencing median OS and PFS. Almost all patients included in the study experienced treatment-related adverse events (TRAEs) of varying degrees of severity, with grade 1-2 adverse events predominating. Lenvatinib/bevacizumab combined with programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) inhibitors and gemcitabine/oxaliplatin (GEMOX) represent an effective and tolerable regimen for advanced BTC in a multicenter retrospective real-world study.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Zhengfeng Zhang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Dazhen Wang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Ze Liu
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China
| | - Changjie Lou
- Department of Gastroenterology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province, China.
| |
Collapse
|
12
|
Singh U, Kokkanti RR, Patnaik S. Beyond chemotherapy: Exploring 5-FU resistance and stemness in colorectal cancer. Eur J Pharmacol 2025; 991:177294. [PMID: 39863147 DOI: 10.1016/j.ejphar.2025.177294] [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: 11/02/2024] [Revised: 12/28/2024] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Colorectal cancer (CRC) remains a significant global health challenge, demanding continuous advancements in treatment strategies. This review explores the complexities of targeting colorectal cancer stem cells (CSCs) and the mechanisms contributing to resistance to 5-fluorouracil (5-FU). The efficacy of 5-FU is enhanced by combination therapies such as FOLFOXIRI and targeted treatments like bevacizumab, cetuximab, and panitumumab, particularly in KRAS wild-type tumors, despite associated toxicity. Biomarkers like thymidylate synthase (TYMS), thymidine phosphorylase (TP), and dihydropyrimidine dehydrogenase (DPD) are crucial for predicting 5-FU efficacy and resistance. Targeting CRC-CSCs remains challenging due to their inherent resistance to conventional therapies, marker variability, and the protective influence of the tumor microenvironment which promotes stemness and survival. Personalized treatment strategies are increasingly essential to address CRC's genetic and phenotypic diversity. Advances in immunotherapy, including immune checkpoint inhibitors and cancer vaccines, along with nanomedicine-based therapies, offer promising targeted drug delivery systems that enhance specificity, reduce toxicity, and provide novel approaches for overcoming resistance mechanisms. Integrating these innovative strategies with traditional therapies may enhance the effectiveness of CRC therapy by addressing the underlying causes of 5-FU resistance in CSCs.
Collapse
Affiliation(s)
- Ursheeta Singh
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, Odisha, India
| | - Rekha Rani Kokkanti
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, Odisha, India
| | - Srinivas Patnaik
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) Deemed to be University, Bhubaneswar, 751024, Odisha, India.
| |
Collapse
|
13
|
Kokori E, Olatunji G, Ogieuhi IJ, Ajayi YI, Akinmoju O, Akinboade A, Irumudomon JG, Omoworare OT, Ezeano C, Adebayo YA, Oyewo O, Aderinto N. The emerging role of Sotorasib plus Panitumumab combination therapy in colorectal cancer treatment. Int J Clin Oncol 2025:10.1007/s10147-025-02736-y. [PMID: 40080361 DOI: 10.1007/s10147-025-02736-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: 07/26/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
Colorectal cancer (CRC) poses a substantial global health challenge, ranking as the third most commonly diagnosed and second most fatal cancer worldwide. With an increasing incidence, particularly in older populations, CRC demands innovative therapeutic approaches to address the limitations of existing treatments. One critical target in CRC is the KRAS gene, which is frequently mutated and implicated in various cancer-related processes. This narrative review explores the promising role of Sotorasib plus Panitumumab combination therapy in CRC treatment. Combining Sotorasib with Panitumumab, an EGFR antagonist, offers a synergistic approach to comprehensively block KRAS and EGFR pathways, potentially overcoming resistance mechanisms observed in monotherapies. The review discusses the evolution of CRC treatment from traditional chemotherapy to the advent of targeted therapies like Bevacizumab and Cetuximab. It highlights the limitations of existing therapies, including resistance and toxicities, emphasising the urgency for innovative approaches. The CodeBreak clinical trials, specifically CodeBreak 101 and CodeBreak 300, provide a focal point for evaluating the efficacy of Sotorasib plus Panitumumab in patients with refractory KRAS G12C-mutated mCRC. Preliminary results demonstrate significant improvements in progression-free survival (PFS) and objective response rates, suggesting a paradigm shift in CRC treatment. The preliminary findings from the CodeBreak 300 trial signify a transformative impact of Sotorasib plus Panitumumab in refractory KRAS G12C-mutated mCRC. With a notable increase in PFS and objective response rates and a well-tolerated safety profile, this combination therapy emerges as a potential new standard of care. The results present an optimistic outlook for patients resistant to conventional therapies.
Collapse
Affiliation(s)
- Emmanuel Kokori
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | - Gbolahan Olatunji
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | - Yusuf Ismaila Ajayi
- Department of Medicine and Surgery, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Olumide Akinmoju
- Department of Medicine, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Adeola Akinboade
- Department of Medicine and Surgery, University of Ilorin, Ilorin, Nigeria
| | | | | | - Chimezirim Ezeano
- Health Science Center, University of North Texas, Fort Worth, Texas, USA
| | | | | | - Nicholas Aderinto
- Department of Medicine and Surgery, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
14
|
Shaham SH, Vij P, Tripathi MK. Advances in Targeted and Chemotherapeutic Strategies for Colorectal Cancer: Current Insights and Future Directions. Biomedicines 2025; 13:642. [PMID: 40149618 PMCID: PMC11940796 DOI: 10.3390/biomedicines13030642] [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: 01/30/2025] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide, necessitating the continuous evolution of therapeutic approaches. Despite advancements in early detection and localized treatments, metastatic colorectal cancer (mCRC) poses significant challenges due to low survival rates and resistance to conventional therapies. This review highlights the current landscape of CRC treatment, focusing on chemotherapy and targeted therapies. Chemotherapeutic agents, including 5-fluorouracil, irinotecan, and oxaliplatin, have significantly improved survival but face limitations such as systemic toxicity and resistance. Targeted therapies, leveraging mechanisms like VEGF, EGFR, and Hedgehog pathway inhibition, offer promising alternatives, minimizing damage to healthy tissues while enhancing therapeutic precision. Furthermore, future directions in CRC treatment include exploring innovative targets such as Wnt/β-catenin, Notch, and TGF-β pathways, alongside IGF/IGF1R inhibition. These emerging strategies aim to address drug resistance and improve patient outcomes. This review emphasizes the importance of integrating molecular insights into drug development, advocating for a more personalized approach to combat CRC's complexity and heterogeneity.
Collapse
Affiliation(s)
- Salique H. Shaham
- Medicine and Oncology ISU, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Puneet Vij
- Department of Pharmaceutical Sciences, St. John’s University, 8000 Utopia Parkway, Queens, New York, NY 11439, USA;
| | - Manish K. Tripathi
- Medicine and Oncology ISU, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
15
|
Egger D, Heger KA, Bolz M, Brinkmann MP, Krepler K, Vecsei-Marlovits PV, Wedrich A, Waldstein SM. Intravitreal therapy-success stories and challenges. Wien Med Wochenschr 2025:10.1007/s10354-024-01070-8. [PMID: 40029473 DOI: 10.1007/s10354-024-01070-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/20/2024] [Indexed: 03/05/2025]
Abstract
Intravitreal injections have revolutionized the treatment of various sight-threatening diseases of the posterior segment of the eye. Initially explored for treatment of bacterial endophthalmitis, intravitreal injections rapidly expanded to combat retinal vascular disease in particular. Especially anti-vascular endothelial growth factor agents have emerged as a cornerstone of intravitreal therapy, targeting neovascular age-related macular degeneration and diabetic macular edema as important examples. Advances continue, with novel therapies such as complement inhibitors now available as treatment for geographic atrophy secondary to non-neovascular age-related macular degeneration, offering hope for a previously untreatable condition. Pioneering approaches such as the port delivery system and intravitreal gene therapy aim to improve treatment efficacy while minimizing patient burden. Despite notable successes, challenges for intravitreal therapies persist, including ocular and systemic complications and high treatment burden. Future research endeavors aim to address these challenges and enhance treatment outcomes. This comprehensive review critically evaluates the efficacy, safety, and cost-effectiveness of intravitreal injections, delving into emerging trends and future directions.
Collapse
Affiliation(s)
- Daniel Egger
- Department of Ophthalmology, Landesklinikum Mistelbach-Gänserndorf, Mistelbach, Austria.
- Karl Landsteiner University of Health Sciences, Krems, Austria.
- Paracelsus Medical University Salzburg, Salzburg, Austria.
| | - Katharina A Heger
- Department of Ophthalmology, Landesklinikum Mistelbach-Gänserndorf, Mistelbach, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Matthias Bolz
- Department of Ophthalmology, Kepler University Clinic, Linz, Austria
- Department of Ophthalmology, Johannes Kepler University, Linz, Austria
| | - Max P Brinkmann
- Department of Ophthalmology, Klinikum Klagenfurt, Klagenfurt, Austria
- Department of Ophthalmology, Universitätsklinikum Schleswig-Holstein, Lübeck, Germany
| | - Katharina Krepler
- Karl Landsteiner Institute for Retinal Research and Imaging, Vienna, Austria
- Department of Ophthalmology, Klinik Landstraße, Vienna, Austria
| | - Pia Veronika Vecsei-Marlovits
- Department of Ophthalmology, Klinik Hietzing, Vienna, Austria
- Karl Landsteiner Institute for Processoptimization and Quality Management in Cataract Surgery, Vienna, Austria
| | - Andreas Wedrich
- Department of Ophthalmology, Medical University of Graz, Graz, Austria
| | - Sebastian M Waldstein
- Department of Ophthalmology, Landesklinikum Mistelbach-Gänserndorf, Mistelbach, Austria
- Karl Landsteiner University of Health Sciences, Krems, Austria
| |
Collapse
|
16
|
Fatemi N, Mirbahari SN, Tierling S, Sanjabi F, Shahrivari S, AmeliMojarad M, Amelimojarad M, Mirzaei Rezaei M, Nobaveh P, Totonchi M, Nazemalhosseini Mojarad E. Emerging Frontiers in Colorectal Cancer Therapy: From Targeted Molecules to Immunomodulatory Breakthroughs and Cell-Based Approaches. Dig Dis Sci 2025; 70:919-942. [PMID: 39869166 PMCID: PMC11919954 DOI: 10.1007/s10620-024-08774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 11/20/2024] [Indexed: 01/28/2025]
Abstract
Colorectal cancer (CRC) is ranked as the second leading cause of cancer-related deaths globally, necessitating urgent advancements in therapeutic approaches. The emergence of groundbreaking therapies, including chimeric antigen receptor-T (CAR-T) cell therapies, oncolytic viruses, and immune checkpoint inhibitors, marks a transformative era in oncology. These innovative modalities, tailored to individual genetic and molecular profiles, hold the promise of significantly enhancing patient outcomes. This comprehensive review explores the latest clinical trials and advancements, encompassing targeted molecular therapies, immunomodulatory agents, and cell-based therapies. By evaluating the strengths, limitations, and potential synergies of these approaches, this research aims to reshape the treatment landscape and improve clinical outcomes for CRC patients, offering new found hope for those who have exhausted conventional options. The culmination of this work is anticipated to pave the way for transformative clinical trials, ushering in a new era of personalized and effective CRC therapy.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Nasim Mirbahari
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| | - Sascha Tierling
- Department of Genetics/Epigenetics, Faculty NT, Life Sciences, Saarland University, Saarbrücken, Germany
| | - Fatemeh Sanjabi
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical, Tehran, Iran
| | - Shabnam Shahrivari
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical, Tehran, Iran
| | - Mandana AmeliMojarad
- Department of Biology, Faculty of Basic Science, Kharrazi University, Tehran, Iran
| | - Melika Amelimojarad
- Department of Biology, Faculty of Basic Science, Kharrazi University, Tehran, Iran
| | - Meygol Mirzaei Rezaei
- School of Advanced Sciences and Technology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Parsa Nobaveh
- School of Advanced Sciences and Technology, Islamic Azad University, Tehran Medical Branch, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, ACECR, Royan Institute for Reproductive Biomedicine, Tehran, Iran
| | - Ehsan Nazemalhosseini Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman St, Chamran Expressway, P.O. Box 19857-17413, Tehran, Iran.
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.
| |
Collapse
|
17
|
Zhang X, Hu J, Fan X, Chen Q, Zheng D, Huang M, Xu Y. The effect of Bevacizumab treatment on the incidence of hypertension in patients with ovarian cancer: a systematic review and meta-analysis. J Oncol Pharm Pract 2025; 31:294-304. [PMID: 39930904 DOI: 10.1177/10781552241307868] [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: 03/14/2025]
Abstract
IntroductionThis study aims to evaluate the effect of bevacizumab treatment on the incidence of hypertension in patients with ovarian cancer.MethodsA comprehensive search of PubMed, Scopus, Embase, Cochrane, Web of Science, and Google Scholar databases was conducted until August 2024. We included only randomized clinical trials that compared ovarian cancer patients treated with Bevacizumab to those treated with other therapies. The primary outcome was the relative risk (RR) of developing hypertension, stratified by grade. Statistical analyses were performed using a random-effects model to account for heterogeneity between studies. Subgroup analyses were conducted based on hypertension severity (grade ≥2 and grade ≥3) and disease stage. Sensitivity analyses and publication bias assessments were also performed.ResultsA total of 11 randomized trials were included, comprising 5212 patients. The meta-analysis revealed that patients receiving Bevacizumab had a significantly higher risk of hypertension compared to controls (RR = 2.91, 95% CI: 1.65-5.16, P = 0.0002). Subgroup analysis showed that the risk of grade ≥2 hypertension was 1.68 times higher (95% CI: 0.92-3.07), and grade ≥3 hypertension was 5.10 times higher (95% CI: 2.46-10.55) in the Bevacizumab group. Sensitivity analysis confirmed the robustness of these findings, and no significant publication bias was detected.ConclusionBevacizumab treatment in ovarian cancer significantly increases the risk of hypertension, particularly severe hypertension (grade ≥3). These findings underscore the need for vigilant blood pressure monitoring and management in patients receiving Bevacizumab to mitigate cardiovascular complications and optimize treatment outcomes.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Pharmaceutical Department, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Jumei Hu
- Department of Gynecology, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Xijing Fan
- Clinical Laboratory, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Qiaoqiao Chen
- Pharmaceutical Department, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Danjun Zheng
- Pharmaceutical Department, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Minjuan Huang
- Pharmaceutical Department, Jinhua Municipal Central Hospital, Jinhua City, China
| | - Yuanqing Xu
- Pharmaceutical Department, Jinhua Municipal Central Hospital, Jinhua City, China
| |
Collapse
|
18
|
Ahn DH, Ridinger M, Cannon TL, Mendelsohn L, Starr JS, Hubbard JM, Kasi A, Barzi A, Samuëlsz E, Karki A, Subramanian RA, Yemane D, Kim R, Wu CC, Croucher PJ, Smeal T, Kabbinavar FF, Lenz HJ. Onvansertib in Combination With Chemotherapy and Bevacizumab in Second-Line Treatment of KRAS-Mutant Metastatic Colorectal Cancer: A Single-Arm, Phase II Trial. J Clin Oncol 2025; 43:840-851. [PMID: 39475591 PMCID: PMC11856007 DOI: 10.1200/jco-24-01266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/09/2024] [Accepted: 09/19/2024] [Indexed: 11/21/2024] Open
Abstract
PURPOSE This phase II study evaluated the efficacy and tolerability of onvansertib, a polo-like kinase 1 (PLK1) inhibitor, in combination with fluorouracil, leucovorin, and irinotecan (FOLFIRI) + bevacizumab for the second-line treatment of KRAS-mutant metastatic colorectal cancer (mCRC). PATIENTS AND METHODS This multicenter, open-label, single-arm study enrolled patients with KRAS-mutated mCRC previously treated with oxaliplatin and fluorouracil with or without bevacizumab. Patients received onvansertib (15 mg/m2 once daily on days 1-5 and 15-19 of a 28-day cycle) and FOLFIRI + bevacizumab (days 1 and 15). The primary end point was the objective response rate (ORR), and secondary endpoints included progression-free survival (PFS), duration of response (DOR), and tolerability. Translational and preclinical studies were conducted in KRAS-mutant CRC. RESULTS Among the 53 patients treated, the confirmed ORR was 26.4% (95% CI, 15.3 to 40.3). The median DOR was 11.7 months (95% CI, 9.4 to not reached). Grade 3/4 adverse events were reported in 62% of patients. A post hoc analysis revealed that patients with no prior bevacizumab treatment had a significantly higher ORR and longer PFS compared with patients with prior bevacizumab treatment: ORR of 76.9% versus 10.0% (odds ratio of 30.0, P < .001) and median PFS of 14.9 months versus 6.6 months (hazard ratio of 0.16, P < .001). Our translational findings support that prior bevacizumab exposure contributes to onvansertib resistance. Preclinically, we showed that onvansertib inhibited the hypoxia pathway and exhibited robust antitumor activity in combination with bevacizumab through the inhibition of angiogenesis. CONCLUSION Onvansertib in combination with FOLFIRI + bevacizumab showed significant activity in the second-line treatment of patients with KRAS-mutant mCRC, particularly in patients with no prior bevacizumab treatment. These findings led to the evaluation of the combination in the first-line setting (ClinicalTrails.gov identifier: NCT06106308).
Collapse
Affiliation(s)
- Daniel H. Ahn
- Division of Medical Oncology, Mayo Clinic, Phoenix, AZ
| | | | | | | | - Jason S. Starr
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL
| | - Joleen M. Hubbard
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Anup Kasi
- Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS
| | - Afsaneh Barzi
- Division of Medical Oncology and Therapeutics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | | | | | | | | | - Roy Kim
- Cardiff Oncology Inc, San Diego, CA
| | | | | | | | | | - Heinz-Josef Lenz
- Division of Oncology, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
19
|
Saadh MJ, Jasim NY, Ahmed MH, Ballal S, Kumar A, Atteri S, Vashishth R, Rizaev J, Alhili A, Jawad MJ, Yazdi F, Salajegheh A, Akhavan-Sigari R. Critical roles of miR-21 in promotions angiogenesis: friend or foe? Clin Exp Med 2025; 25:66. [PMID: 39998742 PMCID: PMC11861128 DOI: 10.1007/s10238-025-01600-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
MiRNAs are small RNA strands that are managed following transcription and are of substantial importance in blood vessel formation. It is essential to oversee the growth, differentiation, death, movement and construction of tubes by angiogenesis-affiliated cells. If miRNAs are not correctly regulated in regard to angiogenesis, it can deteriorate the health and lead to various illnesses, which include cancer, cardiovascular disorder, critical limb ischemia, Crohn's disease, ocular diseases, diabetic microvascular complications, and more. Consequently, it is vital to understand the crucial part that miRNAs play in the development of blood vessels, so we can develop reliable treatment plans for vascular diseases. This write-up will assess the critical role of miR-21/exosomal miR-21 in managing angiogenesis associated with bone growth, wound recovery, and other pathological conditions like tumor growth, ocular illnesses, diabetes, and other diseases connected to formation of blood vessels. Previous investigations have demonstrated that miR-21 is present at higher amounts in certain cancerous cells, and it influences a multitude of genes that moderate the increased creation of blood vessels. Furthermore, studies demonstrated that exosomal miR-21 has the capacity to interact with endothelial cells to foster tumor angiogenesis. For that reason, this review explains the critical importance of miR-21/exosomal miR-21 in managing both healthy and diseased states of angiogenesis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Nisreen Yasir Jasim
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | | | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Abhishek Kumar
- School of Pharmacy-Adarsh Vijendra Institute of Pharmaceutical Sciences, Shobhit University, Gangoh, Uttar Pradesh, 247341, India
- Department of Pharmacy, Arka Jain University, Jamshedpur, Jharkhand, 831001, India
| | - Shikha Atteri
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjheri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Jasur Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, 18, Amir Temur Street, Samarkand, Uzbekistan
| | - Ahmed Alhili
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | | - Farzaneh Yazdi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| | | | - Reza Akhavan-Sigari
- Dr. Schneiderhan GmbH and ISAR Klinikum, Munich, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw, Management University Warsaw, Warsaw, Poland
| |
Collapse
|
20
|
Mahmoud A, Choi PH, Sukhwa C, Pintar J, Walch H, Zhao N, Bermeo J, Chung S, Raghavan M, Bapat S, Jiang Q, Karagkounis G, Meredith J, Giarrizzo M, Firat C, Cercek A, Foote MB, Schultz N, Chatila WK, Nash GM, Shia J, Sanchez-Vega F, Larson S, Dar AC, Rosen N, Ganesh K. Paired primary-metastasis patient-derived organoids and mouse models identify phenotypic evolution and druggable dependencies of peritoneal metastasis from appendiceal cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638725. [PMID: 40027618 PMCID: PMC11870485 DOI: 10.1101/2025.02.17.638725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Peritoneal carcinomatosis is a common yet deadly manifestation of gastrointestinal cancers, with few effective treatments. To identify targetable determinants of peritoneal metastasis, we focused on appendiceal adenocarcinoma (AC), a gastrointestinal cancer that metastasizes almost exclusively to the peritoneum. Current treatments are extrapolated from colorectal cancer (CRC), yet AC has distinct genomic alterations, mucinous morphology and peritoneum restricted metastatic pattern. Further, no stable preclinical models of AC exist, limiting drug discovery and representing an unmet clinical need. We establish a first-in-class stable biobank of 16 long-term cultured AC patient-derived organoids (PDOs), including 3 matched, simultaneously resected primary AC-peritoneal carcinomatosis (AC-PC) pairs. By enriching for cancer cells, AC PDOs enable accurate genomic characterization relative to paucicellular AC tissue. We establish an organoid orthotopic intraperitoneal xenograft model that recapitulates diffuse peritoneal carcinomatosis and show that PC-organoids retain increased metastatic capacity, decreased growth factor dependency and sensitivity to standard of care chemotherapy relative to matched primary AC organoids. Single cell profiling of AC-PC pairs reveals dedifferentiation from mucinous differentiated states in primary AC into intestinal stem cell and fetal progenitor states in AC-PC, with upregulation of oncogenic signaling pathways. Through hypothesis-driven drug testing, we identify KRAS MULTI -ON inhibitor RMC-7977 and Wnt-targeting tyrosine kinase inhibitor WNTinib as novel, clinically actionable strategies to target AC-PC more effectively.
Collapse
|
21
|
Xie Y, Tang S, Qin Z, Yang C. Clinical Progress of Fruquintinib in Colorectal Cancer: An Overview. Pharmaceuticals (Basel) 2025; 18:280. [PMID: 40006092 PMCID: PMC11859084 DOI: 10.3390/ph18020280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/05/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies worldwide, with high morbidity and mortality rates. Conventional treatments, including surgery, radiotherapy, and chemotherapy, have limited effects on advanced and metastatic CRC (mCRC). Fruquintinib, a novel and highly selective vascular endothelial growth factor receptor (VEGFR) inhibitor, has shown significant efficacy and tolerance in treating mCRC. The FRESCO and FRESCO-2 trials demonstrated that fruquintinib significantly prolongs progression-free survival and the overall survival of refractory mCRC patients, establishing it as the standard third-line treatment strategy for mCRC. In addition, the combination of fruquintinib with other anticancer drugs and immune checkpoint inhibitors demonstrated potential for enhanced efficacy, which warrants further exploration. In this review, we aimed to systematically summarize the current knowledge about the pharmacological mechanisms, pharmacokinetic characteristics, adverse events, and corresponding treatment options of fruquintinib and provide an update on the clinical trials related to fruquintinib in CRC by conducting a comprehensive literature search of PubMed and consulting the relevant clinical trials via ClinicalTrials.gov and the ChiCTR website, aiming to offer new insights into the role of fruquintinib in the comprehensive treatment of CRC.
Collapse
Affiliation(s)
- Yejie Xie
- The Second Clinical School of Wuhan University, Wuhan 430071, China; (Y.X.)
| | - Shu Tang
- Department of Anesthesia and Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ziheng Qin
- The Second Clinical School of Wuhan University, Wuhan 430071, China; (Y.X.)
| | - Chaogang Yang
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Hubei Key Laboratory of Tumour Biological Behaviours, Hubei Cancer Clinical Study Centre, The Clinical Medical Research Centre of Peritoneal Cancer of Wuhan, Wuhan 430071, China
| |
Collapse
|
22
|
Vonica RC, Morgovan C, Butuca A, Pumnea M, Cipaian RC, Frum A, Dobrea CM, Vonica-Tincu AL, Pacnejer AM, Batar F, Vornicu V, Ghibu S, Gligor FG. Real-World Evidence of Bevacizumab and Panitumumab Drug Resistance and Drug Ineffectiveness from EudraVigilance Database. Cancers (Basel) 2025; 17:663. [PMID: 40002260 PMCID: PMC11853327 DOI: 10.3390/cancers17040663] [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: 01/17/2025] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in the world, with an average 5-year overall survival (OS) rate of approximately 60% [...].
Collapse
Affiliation(s)
- Razvan Constantin Vonica
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Claudiu Morgovan
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Anca Butuca
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Manuela Pumnea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Remus Calin Cipaian
- Clinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania;
- County Clinical Emergency Hospital of Sibiu, 2-4 Corneliu Coposu Str., 550245 Sibiu, Romania
| | - Adina Frum
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Carmen Maximiliana Dobrea
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Andreea Loredana Vonica-Tincu
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Aliteia-Maria Pacnejer
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
- Department of Toxicology, Drug Industry, Management and Legislation, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania
| | - Florina Batar
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| | - Vlad Vornicu
- Department IX Surgery, Discipline of Oncology, Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timisoara, Romania;
| | - Steliana Ghibu
- Department of Pharmacology, Physiology and Pathophysiology, Faculty of Pharmacy, “Iuliu Haţieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Felicia Gabriela Gligor
- Preclinical Department, Faculty of Medicine, “Lucian Blaga” University of Sibiu, 550169 Sibiu, Romania; (R.C.V.); (M.P.); (A.F.); (C.M.D.); (A.L.V.-T.); (A.-M.P.); (F.B.); (F.G.G.)
| |
Collapse
|
23
|
Guo H, Miao L, Yu C. The efficacy of targeted therapy and/or immunotherapy with or without chemotherapy in patients with colorectal cancer: A network meta-analysis. Eur J Pharmacol 2025; 988:177219. [PMID: 39716565 DOI: 10.1016/j.ejphar.2024.177219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/17/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND The use of targeted drugs and immunotherapy has significantly impacted the treatment of Colorectal Cancer. However, horizontal comparison among various regimens is extremely rare. Therefore, we evaluated the survival efficacy of multiple treatment regimens of targeted therapy and/or immunotherapy with or without chemotherapy in patients with Colorectal Cancer. METHODS A systematic search was conducted in PubMed, EMBASE, and Cochrane databases, covering the period from the establishment of the databases to October 29, 2024. To obtain articles that met the inclusion and exclusion criteria and contained the required data for conducting a network meta-analysis (NMA). The NMA evaluated overall survival (OS) and progression-free survival (PFS). RESULTS A total of 90 studies were identified, comprising a sample size of 33,167 subjects. In terms of PFS, compared with simple chemotherapy strategies, most of the other single or combined strategies are significantly effective, among which targeted therapy strategies have more advantages. Encorafenib + Binimetinib + Cetuximab (ENC-BIN-CET) shows significant benefits in all comparisons except when compared with Chemotherapy + Cetuximab + Dalotuzumab (Chemo-CET-DAL), Encorafenib + Cetuximab (ENC-CET), and Panitumumab + Sotorasib (PAN-SOT). The ENC-CET and PAN-SOT targeted strategies also show significant benefits. Pembrolizumab (PEM) monotherapy has advantages over all others except when it is not superior to some targeted strategies. Chemotherapy + Bevacizumab + Atezolizumab is only inferior to some strategies. In terms of OS, the combinations of Chemotherapy + Bevacizumab, ENC-CET, Chemotherapy + Panitumumab, and ENC-BIN-CET are superior to simple chemotherapy regimens. ENC-BIN-CET shows OS benefits in all comparisons except some. ENC-CET significantly improves OS in most cases, and PEM also significantly improves OS in some regimens. In the probability ranking of OS and PFS, ENC-BIN-CET has the best effect, followed by ENC-CET. CONCLUSIONS In conclusion, pembrolizumab is still effective in prolonging survival. Dual- and triple-drug targeted strategies are the best in terms of OS and PFS, and the combination of targeted immunotherapy and chemotherapy also works. However, not all combinations are beneficial. As targeted drugs play an active role, specific drugs for colorectal cancer regimens should be carefully selected.
Collapse
Affiliation(s)
- Haoyan Guo
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, No.16, Guicheng South Fifth Road, Foshan, Guangdong, 528200, China; Jinan University, Guangzhou, 510632, China
| | - Longjie Miao
- Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, Guangdong, 518104, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chengdong Yu
- Nanhai Hospital of Traditional Chinese Medicine, Jinan University, No.16, Guicheng South Fifth Road, Foshan, Guangdong, 528200, China; Jinan University, Guangzhou, 510632, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
24
|
Li X, Zhu D, Zhao B, Li Q, Jin P. Alternative splicing: Therapeutic target for vasculopathy in diabetic complications. Life Sci 2025; 362:123331. [PMID: 39734014 DOI: 10.1016/j.lfs.2024.123331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 12/31/2024]
Abstract
It is becoming increasingly evident that diabetic vascular complications seriously threaten human health. The most prevalent microvascular complications include kidney disease, retinal disease, cardiovascular diseases and amputation. Conventional treatments can only relieve the progression of the diseases, and is no longer appropriate for the long-term management of diabetic patients. Exploring a novel therapeutic regimens and improvements in management of Diabetic Complications is required. Alternative splicing has been found to play a crucial role in the occurrence and treatment of diseases, including the destruction and generation of blood vessels in diabetes. Alternative splicing is an important factor in the high complexity of multicellular eukaryotic transcriptome, and angiogenesis, which is an important process controlled by alternative splicing mechanism. This review mainly introduces the current understanding of alternative splicing and the role that alternative splicing plays in the diabetic complications, with a special focus on vascular system. In this study, we summarized alternative splicing in relation to diabetes complications and the pathogenesis of diabetic vasculopathy. It discussed potential treatment strategies for correcting aberrant splicing and suggested novel approaches for addressing diabetes complications.
Collapse
Affiliation(s)
- Xiaoyue Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Xuzhou Medical University, Xuzhou, China
| | - Dong Zhu
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Xuzhou Medical University, Xuzhou, China
| | - Bingkun Zhao
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Qiang Li
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Peisheng Jin
- Department of Plastic Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
25
|
Hughes H, Jajodia A, Soyer P, Mellnick V, Patlas MN. Bowel Emergencies in Patients With Cancer. Can Assoc Radiol J 2025; 76:76-86. [PMID: 38721789 DOI: 10.1177/08465371241252035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024] Open
Abstract
Cancer is the second most common cause of death worldwide. Bowel emergencies in patients with cancer are becoming increasingly more prevalent due to advances in cancer therapy and longer overall patient survival. When these patients present acutely, they are often frail and may have pre-existing co-morbidities. This article discusses the imaging features of bowel emergencies commonly encountered in oncological patients in clinical practice. These include chemotherapy related colitis, neutropenia enterocolitis and typhlitis, toxic megacolon, bowel perforation, malignant bowel obstruction and gastrointestinal haemorrhage. The radiologist plays a key role in identifying these oncological emergencies and guiding further management.
Collapse
Affiliation(s)
- Hannah Hughes
- Department of Radiology, St. Vincent's University Hospital, Dublin, Ireland
| | - Ankush Jajodia
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - Philippe Soyer
- Department of Radiology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
- Faculté de Médecine, Université Paris Cité, Paris, France
| | - Vincent Mellnick
- Department of Radiology, Mallinckrodt Institute of Radiology, St Louis, MO, USA
| | - Michael N Patlas
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
26
|
Alexander HR, Devi-Chou V. Hepatic Perfusion for Diffuse Metastatic Cancer to the Liver: Open and Percutaneous Techniques. Hematol Oncol Clin North Am 2025; 39:177-190. [PMID: 39510672 DOI: 10.1016/j.hoc.2024.08.007] [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: 11/15/2024]
Abstract
The management of patients with diffuse liver metastases remains a significant clinical challenge. In many cancer patients, metastatic disease may be isolated to the liver or the liver may be the dominant site of progressive metastatic cancer. In this setting, progression of disease in the liver generally is the most significant cause of morbidity and mortality.
Collapse
Affiliation(s)
- H Richard Alexander
- Department of Surgery, Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, 195 Little Albany Street, Room 2009, New Brunswick, NJ 08901, USA.
| | - Virginia Devi-Chou
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
27
|
Li J, Wang D, Duan Q, Su N, Li X, Qiu H. The efficacy of anti-angiogenic drugs in gastric-type endocervical adenocarcinoma: A retrospective study. J Obstet Gynaecol Res 2025; 51:e16247. [PMID: 39988602 DOI: 10.1111/jog.16247] [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: 09/28/2024] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
OBJECTIVES Gastric-type endocervical adenocarcinoma (GEA) is a rare malignant tumor that is not associated with high-risk HPV infection, known for its high invasiveness and resistance to current treatments. This study assessed the effectiveness of anti-angiogenic regimens in real-world GEA patients. METHODS Patients with GEA were enrolled between February 2012 and March 2023, and their clinicopathological characteristics were collected from their medical records. The patients were categorized into groups based on whether they received anti-angiogenic treatments or not. Survival analysis was conducted using the Kaplan-Meier method. RESULTS A total of 43 GEA patients were enrolled in this study, with 23 cases who received anti-angiogenic drugs (nine received them as the primary treatment, 12 as first-line therapy after recurrence/metastasis, and two as second-line therapy) as the observation group. The other 20 patients who received similar treatments without the anti-angiogenic regimens serve as the control group. Compared to the control group, the addition of anti-angiogenic drugs as the primary treatment mildly extended progression-free survival (PFS) while not being statistically significant (16 months vs 11 months, p = 0.744). The negative results were also observed in 12 patients who started anti-angiogenic therapy as first-line therapy after recurrence/metastasis (8.5 months vs 9 months, p = 0.518). As for the overall survival (OS), no benefits were detected in either patients who started the anti-angiogenic therapy as primary or subsequent treatments (p = 0.499 and 0.450, respectively). CONCLUSION We firstly evaluated the efficacy of anti-angiogenic drugs in treating patients with GEA. Although with a small sample size, our preliminary results clearly proposed that the anti-angiogenic therapy failed in suppressing tumors and should not be a preferred choice for GEA. As a much rarer tumor without standard treatments, we herein warned of a pitfall for gynecologic oncologists when facing this malignancy.
Collapse
Affiliation(s)
- Jing Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Dian Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Qing Duan
- Department of Gynecologic Oncology, Anyang Tumor Hospital, Anyang, Henan Province, China
| | - Ning Su
- Department of Gynecologic Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Xiufang Li
- Department of Gynecologic Oncology, Anyang Tumor Hospital, Anyang, Henan Province, China
| | - Haifeng Qiu
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
28
|
Ji P, Chen T, Li C, Zhang J, Li X, Zhu H. Comprehensive review of signaling pathways and therapeutic targets in gastrointestinal cancers. Crit Rev Oncol Hematol 2025; 206:104586. [PMID: 39653094 DOI: 10.1016/j.critrevonc.2024.104586] [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: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024] Open
Abstract
Targeted therapy, the milestone in the development of human medicine, originated in 2004 when the FDA approved the first targeted agent bevacizumab for colorectal cancer treatment. This new development has resulted from drug developers moving beyond traditional chemotherapy, and several trials have popped up in the last two decades with an unprecedented speed. Specifically, EGF/EGFR, VEGF/VEGFR, HGF/c-MET, and Claudin 18.2 therapeutic targets have been developed in recent years. Some targets previously thought to be undruggable are now being newly explored, such as the RAS site. However, the efficacy of targeted therapy is extremely variable, especially with the emergence of new drugs and the innovative use of traditional targets for other tumors in recent years. Accordingly, this review provides an overview of the major signaling pathway mechanisms and recent advances in targeted therapy for gastrointestinal cancers, as well as future perspectives.
Collapse
Affiliation(s)
- Pengfei Ji
- Department of Thoracic Surgery, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China
| | - Tingting Chen
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Chao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Jinyuan Zhang
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Xiao Li
- The Second Clinical Medical College, Lanzhou University, No. 199 DongGang West Road, Lanzhou, Gansu 730000, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, No. 37 GuoXue Xiang, Chengdu, Sichuan 610041, China.
| |
Collapse
|
29
|
Taniguchi H, Uehara K, Ishikawa T, Okochi O, Akazawa N, Okuda H, Hasegawa H, Shiozawa M, Kataoka M, Satake H, Shimura T, Kondoh C, Kuramochi H, Matsumoto T, Takegawa N, Yamaguchi T, Nagase M, Nakamura M, Takano N, Fujita H, Watanabe T, Nishina T, Sakamoto Y, Moriwaki T, Ohori H, Nakanishi M, Kito Y, Utsunomiya S, Ishikawa T, Manaka D, Matsuoka H, Suto T, Arai T, Shinzaki S, Funakoshi T, Nakayama G, Negoro Y, Tsuji Y, Makiyama A, Takuma K, Arimoto A, Shinozaki K, Mishima A, Masuishi T. BRAF V600E and Non-V600E Mutations in RAS Wild-Type Metastatic Colorectal Cancer: Prognostic and Therapeutic Insights from a Nationwide, Multicenter, Observational Study (J-BROS). Cancers (Basel) 2025; 17:399. [PMID: 39941768 PMCID: PMC11815755 DOI: 10.3390/cancers17030399] [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: 12/29/2024] [Revised: 01/19/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES BRAF mutations occur in 5-10% of metastatic colorectal cancer (mCRC) cases, but their implications for prognosis and optimal treatment remain unclear. METHODS This multicenter, prospective observational study analyzed 377 RAS wild-type cases from 511 patients across 32 centers, using PCR-based methods. RESULTS BRAF mutations were identified in 21% (79/377) of cases, predominantly V600E (89.9%) with a minority of non-V600E (10.1%). Microsatellite instability (MSI) testing revealed MSI-high in 11.3%, exclusively among V600E cases. V600E mutations were linked to right-sided tumors, poor differentiation, and elevated CA19-9 levels. Median survival was significantly lower in V600E cases compared to BRAF wild-type (12.4 vs. 37.5 months, HR 3.25, p < 0.001) and marginally lower non-V600E cases (12.4 vs. 34.7 months, HR 0.61, p = 0.057). Chemotherapy regimens (doublet vs. triplet) and targeted treatments (bevacizumab vs. anti-EGFR) showed no significant survival differences in V600E patients. Similarly, RAS/BRAF wild-type patients had comparable survival with bevacizumab versus anti-EGFR, even for left-sided tumors. CONCLUSIONS These findings highlight distinct clinical and prognostic profiles for BRAF V600E and non-V600E mutations, while treatment choice appears to have limited impact on survival in these subgroups or RAS/BRAF wild-type cases.
Collapse
Affiliation(s)
- Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan
| | - Kay Uehara
- Department of Gastroenterological Surgery, Nagoya University Hospital, Nagoya 466-8560, Japan
- Department of Gastroenterological Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| | - Toshiaki Ishikawa
- Department of Specialized Surgeries, Institute of Science Tokyo, Tokyo 113-8519, Japan
- Department of Medical Oncology, Juntendo University, Tokyo 113-8431, Japan
| | - Osamu Okochi
- Department of Surgery, Tosei General Hospital, Seto 489-8642, Japan
| | - Naoya Akazawa
- Department of Gastroenterological Surgery, Sendai City Medical Center Sendai Open Hospital, Sendai 983-0824, Japan
| | - Hiroyuki Okuda
- Department of Clinical Oncology, Keiyukai Sapporo Hospital, Sapporo 003-0026, Japan
| | - Hiroko Hasegawa
- Department of Gastroenterology and Hepatology, NHO Osaka National Hospital, Osaka 540-0006, Japan
| | - Manabu Shiozawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama 241-0815, Japan
| | - Masato Kataoka
- Department of Surgery, NHO Nagoya Medical Center, Nagoya 460-0001, Japan
| | - Hironaga Satake
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe 650-0047, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku 783-8505, Japan
| | - Takaya Shimura
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8602, Japan
| | - Chihiro Kondoh
- Department of Medical Oncology, Toranomon Hospital, Tokyo 105-8470, Japan
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan
| | - Hidekazu Kuramochi
- Department of Chemotherapy, Tokyo Women’s Medical University Yachiyo Medical Center, Yachiyo 276-8524, Japan
- Department of Medical Oncology, NTT Medical Center Tokyo, Tokyo 141-8625, Japan
| | - Toshihiko Matsumoto
- Department of Internal medicine, Himeji Red Cross Hospital, Himeji 670-8540, Japan
- Department of Medical Oncology, Ichinomiyanishi Hospital, Ichinomiya 494-0001, Japan
| | - Naoki Takegawa
- Department of Gastroenterology, Hyogo Cancer Center, Akashi 673-8558, Japan
| | - Toshifumi Yamaguchi
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki 569-0801, Japan
| | - Michitaka Nagase
- Department of Medical Oncology, Saku Central Hospital Advanced Care Center, Saku 385-0051, Japan
| | - Masato Nakamura
- Aizawa Comprehensive Cancer Center, Aizawa Hospital, Matsumoto 390-8510, Japan
| | - Nao Takano
- Department of Surgery, Tokai Central Hospital, Kagamihara 504-8601, Japan
- Department of Advanced Medicine, Nagoya University Hospital, Nagoya 466-8560, Japan
| | - Hideto Fujita
- Department of General and Digestive surgery, Kanazawa Medical University, Uchinadamachi 920-0293, Japan
| | - Takanori Watanabe
- Department of Surgery, Japanese Red Cross Society Himeji Hospital, Himeji 670-8540, Japan
- Department of Surgery, Tokushima Municipal Hospital, Tokushima 770-0812, Japan
| | - Tomohiro Nishina
- Department of Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama 791-0245, Japan
| | - Yasuhiro Sakamoto
- Department of Medical Oncology, Osaki Citizen Hospital, Osaki 989-6183, Japan
| | - Toshikazu Moriwaki
- Department of Gastroenterology, University of Tsukuba, Tsukuba 305-8576, Japan
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Kurashiki 710-8602, Japan
| | - Hisatsugu Ohori
- Department of Medical Oncology, Ishinomaki Red Cross Hospital, Ishinomaki 986-8522, Japan
| | - Masayoshi Nakanishi
- Department of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Surgery, Matsushita Memorial Hospital, Moriguchi 570-8540, Japan
| | - Yosuke Kito
- Department of Medical Oncology, Ishikawa Prefectural Central Hospital, Kanazawa 920-8530, Japan
| | - Setsuo Utsunomiya
- Department of Clinical Oncology, Kainan Hospital, Yatomi 498-8502, Japan
| | - Takeshi Ishikawa
- Outpatient Oncology Unit, Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Dai Manaka
- Department of Surgery, Kyoto Katsura Hospital, Kyoto 615-8256, Japan
| | - Hiroshi Matsuoka
- Department of Surgery, Fujita Health University, Toyoake 470-1192, Japan
| | - Takeshi Suto
- Department of Gastroenterological Surgery, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| | - Toshiyuki Arai
- Department of Surgery, Anjo Kosei Hospital, Anjo 446-8602, Japan
| | - Shinichiro Shinzaki
- Department of Gastroenterology, School of Medicine, Hyogo Medical University, Nishinomiya 663-8501, Japan
| | - Tohru Funakoshi
- Department of Surgery, Asahikawa Kosei General Hospital, Asahikawa 078-8211, Japan
| | - Goro Nakayama
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan
| | - Yuji Negoro
- Department of Oncological Medicine, Kochi Health Sciences Center, Kochi 781-8555, Japan
| | - Yasushi Tsuji
- Department of Medical Oncology, Tonan Hospital, Sapporo 060-0004, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Kitakyushyu 806-0034, Japan
- Cancer Center, Gifu University Hospital, Gifu 501-1194, Japan
| | - Kunio Takuma
- Department of Surgery, Tokyo Metropolitan Tama Medical Center, Fuchu 183-8524, Japan
| | - Atsuki Arimoto
- Department of General Surgery, Toyohashi Municipal Hospital, Toyohashi 441-8570, Japan
| | - Katsunori Shinozaki
- Division of Clinical Oncology, Hiroshima Prefectural Hospital, Hiroshima 734-8530, Japan
| | - Ayako Mishima
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan
| | - Toshiki Masuishi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya 464-8681, Japan
| |
Collapse
|
30
|
Liu X. Effects of Shenmai Injection on proliferation, migration, invasion and angiogenesis of colorectal carcinoma vascular endothelial cells. Medicine (Baltimore) 2025; 104:e41307. [PMID: 39833060 PMCID: PMC11749673 DOI: 10.1097/md.0000000000041307] [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: 11/29/2023] [Accepted: 10/24/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Colorectal cancer, being 1 of the most significant malignant tumors globally, poses a substantial risk to human health. Unfortunately, its 5-year survival rate stands at a mere 65%. There remains an urgent need for the development of novel treatments to combat this detrimental malignancy effectively. The Shenmai Injection (SMI) is a Chinese medicine that has been proven to have significant clinical efficacy in the treatment of cardiovascular diseases. This study aimed to examine the impact of SMI on the proliferation, migration, invasion, and angiogenesis of tumor-derived endothelial cells (Td-EC). METHODS Human umbilical vein endothelial cells (HUVEC) induced Td-EC, and HUVEC were treated with conditioned media from the human colorectal carcinoma cells (HCT116). The effects of HCT116 on the proliferation, migration, and invasion of hepatocellular carcinoma cells after treatment of SMI were observed by MTS assay and Transwell techniques. Additionally, an angiogenesis experiment was used to investigate Td-EC tube formation capacity. RESULTS SMI had a significant inhibiting effect on the proliferation, migration, and invasion of HCT116. SMI was also able to inhibit the angiogenesis of Td-EC. Notably, SMI did not have any effect on the normal endothelium. CONCLUSION SMI has obvious antiproliferation, migration, infiltration, and neogenesis effects on HCT116.
Collapse
Affiliation(s)
- Xiangyu Liu
- Lanzhou University Second Hospital, Chengguan District, Lanzhou, China
| |
Collapse
|
31
|
Zhou H, Wang Y, Wang F, Meng R, Yu Y, Han S, Zhang Y, Wu Y, Liu X. Assessing cross-country inequalities in global burden of gastrointestinal cancers: slope and concentration index approach. Discov Oncol 2025; 16:41. [PMID: 39806164 PMCID: PMC11729605 DOI: 10.1007/s12672-025-01762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
PURPOSE New cases and deaths of gastrointestinal cancers are predicted to increase significantly by 2040. This study aims to explore cross-country inequalities and trends in global burdens of colon and rectum cancer (CRC), esophageal cancer (EC) and gastric cancer (GC). METHODS Data from the Global Burden of Diseases Study 2019 were analyzed to examine trends in disability-adjusted life-years (DALYs) for three gastrointestinal cancers with estimated annual percentage change (EAPC) and Joinpoint analysis. Inequality in their DALYs rates was assessed with the slope index of inequality and the concentration index, based on the Socio-Demographic Index (SDI). RESULTS From 1990 to 2019, the age standardized DALYs rate of CRC decreased in these countries from high and high-middle SDI regions, with the EAPC values of - 1.018% and - 0.161%, respectively, but increased among low, low-middle and middle SDI regions (EAPC = 1.035%, 0.926% and 0.406%, respectively). The age standardized DALYs rates of EC and GC decreased in all SDI regions. Moreover, the slope index changed from 358.42 (95% confidence interval 343.28 to 370.49) to 245.13 (217.47 to 271.24) for CRC, from - 63.88 (- 87.48 to - 48.28) to - 1.36 (- 32.44 to 25.87) for EC, and from 126.37 (101.97 to 146.47) to 58.04 (20.54 to 96.12) for GC. The concentration index for CRC moved from 29.56 (28.99 to 29.84) to 23.90 (23.19 to 24.26), from - 9.47 (- 10.30 to - 9.24) to - 14.64 (- 15.35 to - 14.24) for EC, and from 8.44 (7.85 to 8.72) to - 6.42 (- 7.65 to - 6.12) for GC. CONCLUSION This study suggests strong heterogeneity in global DALYs for gastrointestinal cancers across different SDI regions. Higher SDI regions faced a greater burden of CRC, while the burdens of EC and GC were more prevalent in lower SDI regions.
Collapse
Affiliation(s)
- Haoyun Zhou
- Global Health Research Division, Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yongbo Wang
- Center for Evidence-Based and Translational Medicine, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Fang Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, China
| | - Runtang Meng
- Department of Preventive Medicine, School of Public Health, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yong Yu
- School of Public Health and Management, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Su Han
- Global Health Research Division, Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yu Zhang
- Global Health Research Division, Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Yu Wu
- Global Health Research Division, Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China
| | - Xiaoxue Liu
- Global Health Research Division, Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
32
|
Arabi S, Fadaee M, Kazemi T, Rahmani M. Advancements in colorectal cancer immunotherapy: from CAR-T cells to exosome-based therapies. J Drug Target 2025:1-12. [PMID: 39754507 DOI: 10.1080/1061186x.2024.2449482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Colorectal cancer (CRC) continues to be a major worldwide health issue, with elevated death rates linked to late stages of the illness. Immunotherapy has made significant progress in developing effective techniques to improve the immune system's capacity to identify and eradicate cancerous cells. This study examines the most recent advancements in CAR-T cell treatment and exosome-based immunotherapy for CRC. CAR-T cell therapy, although effective in treating blood cancers, encounters obstacles when used against solid tumours such as CRC. These obstacles include the presence of an immunosuppressive tumour microenvironment and a scarcity of tumour-specific antigens. Nevertheless, novel strategies like dual-receptor CAR-T cells and combination therapy involving cytokines have demonstrated promise in surmounting these obstacles. Exosome-based immunotherapy is a promising approach for targeted delivery of therapeutic drugs to tumour cells, with high specificity and minimal off-target effects. However, there are still obstacles to overcome in the field, such as resistance to treatment, adverse effects associated with the immune system, and the necessity for more individualised methods. The current research is focused on enhancing these therapies, enhancing the results for patients, and ultimately incorporating these innovative immunotherapeutic approaches into the standard treatment protocols for CRC.
Collapse
Affiliation(s)
- Sepideh Arabi
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammadreza Rahmani
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
33
|
Tang YC, Ou JJ, Hsu SC, Huang CH, Lin LM, Chang HH, Wang YH, Huang ZT, Sun M, Liu KJ, Hung YM, Lai CY, Shih C, Chen CT, Chang JY, Hsieh HP, Jiaang WT, Kuo CC. Advancing precision therapy for colorectal cancer: Developing clinical indications for multi-target kinase inhibitor BPR1J481 using patient-derived xenograft models. Pharmacol Res 2025; 211:107556. [PMID: 39709137 DOI: 10.1016/j.phrs.2024.107556] [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: 06/17/2024] [Revised: 12/04/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
The large and rapid increase in the incidence and mortality of colorectal cancer (CRC) demonstrates the urgent need for new drugs with higher efficacy to treat CRC. However, the lack of applicable and reliable preclinical models significantly hinders the progress of drug development. Patient-derived xenograft (PDX) models are currently considered reliable in vivo preclinical models for predicting drug efficacy in cancer patients. This study successfully uses the CRC PDX model to develop clinical indications for the new multi-target kinase inhibitor BPR1J481 and demonstrated the anti-cancer mechanism and competitive advantages of this drug candidate. The results demonstrate that BPR1J481 exhibits significant anticancer efficacy by inducing apoptosis in CRC PDX tumor tissues and corresponding PDX-derived CRC cells. Through kinase competitive binding and kinase activity assays, we discover that BPR1J481 effectively inhibits SRC kinase activity by directly binding to its active site. The reduction in SRC phosphorylation observed in CRC PDX tumor tissues and derived cells upon treatment with BPR1J481 further validates its inhibitory potential. Furthermore, the decrease in viable cells after SRC knockout and the poorer prognosis observed in patients with higher SRC expression, emphasizes the critical significance and clinical relevance of SRC in CRC. Additionally, BPR1J481 exhibits robust anti-angiogenic effects by suppressing VEGF- and PDGF-induced endothelial cell proliferation, migration, and capillary-like tube formation through inhibition of VEGFR2 and PDGFRβ phosphorylation. Remarkably, BPR1J481 appears to demonstrate greater efficacy against CRC compared to regorafenib. These findings highlight the therapeutic potential of BPR1J481 for patients with CRC.
Collapse
Affiliation(s)
- Ya-Chu Tang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jing-Jim Ou
- Department of Surgery, Chang Bing Show Chwan Memorial Hospital, Changhua County 505029, Taiwan
| | - Shu-Ching Hsu
- Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chih-Hsiang Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Li-Mei Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Yi-Hsin Wang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Zih-Ting Huang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Manwu Sun
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Yi-Mei Hung
- National Institute of Cancer Research, National Health Research Institutes, Tainan City 704016, Taiwan
| | - Chi-Yun Lai
- Pathology Core Laboratory, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chuan Shih
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Taipei Medical University Hospital, College of Medicine, Taipei Medical University, Taipei City 110301, Taiwan; Taipei Cancer Center, Taiwan; TMU Research Center of Cancer Translational Medicine, 110301, Taiwan
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Weir-Torn Jiaang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| |
Collapse
|
34
|
Li Y, Wu W, Yao J, Wang S, Wu X, Yan J. Patient-Derived Tumor Organoids: A Platform for Precision Therapy of Colorectal Cancer. Cell Transplant 2025; 34:9636897251314645. [PMID: 39953837 PMCID: PMC11829288 DOI: 10.1177/09636897251314645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/12/2024] [Accepted: 12/29/2024] [Indexed: 02/17/2025] Open
Abstract
Colorectal cancer (CRC) represents a significant cause of cancer-related mortality on a global scale. It is a highly heterogeneous cancer, and the response of patients to homogeneous drug therapy varies considerably. Patient-derived tumor organoids (PDTOs) represent an optimal preclinical model for cancer research. A substantial body of evidence from numerous studies has demonstrated that PDTOs can accurately predict a patient's response to different drug treatments. This article outlines the utilization of PDTOs in the management of CRC across a range of therapeutic contexts, including postoperative adjuvant chemotherapy, palliative chemotherapy, neoadjuvant chemoradiotherapy, targeted therapy, third-line and follow-up treatment, and the treatment of elderly patients. This article delineates the manner in which PDTOs can inform therapeutic decisions at all stages of CRC, thereby assisting clinicians in selecting treatment options and reducing the risk of toxicity and resistance associated with clinical drugs. Moreover, it identifies shortcomings of existing PDTOs, including the absence of consistent criteria for assessing drug sensitivity tests, the lack of vascular and tumor microenvironment models, and the high cost of the technology. In conclusion, despite their inherent limitations, PDTOs offer several advantages, including rapid culture, a high success rate, high consistency, and high throughput, which can be employed as a personalized treatment option for CRC. The use of PDTOs in CRC allows for the prediction of responses to different treatment modalities at various stages of disease progression. This has the potential to reduce adverse drug reactions and the emergence of resistance associated with clinical drugs, facilitate evidence-based clinical decision-making, and guide CRC patients in the selection of personalized medications, thereby advancing the individualized treatment of CRC.
Collapse
Affiliation(s)
- Yiran Li
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Wei Wu
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Jiaxin Yao
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Suidong Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
| | - Xiufeng Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, P.R. China
| | - Jun Yan
- Department of General Surgery, Guangdong Provincial Key Laboratory of Precision Medicine for Gastrointestinal Tumor, Nanfang Hospital, Southern Medical University, Guangzhou, P.R. China
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, P.R. China
| |
Collapse
|
35
|
Şahinbaş M, Çerik İB, Yalınbaş Yeter D. Investigation of the effect of intravitreal bevacizumab treatment on left heart function using speckle tracking echocardiography. Rev Port Cardiol 2025; 44:27-35. [PMID: 39216529 DOI: 10.1016/j.repc.2024.07.006] [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: 03/14/2024] [Revised: 06/01/2024] [Accepted: 07/07/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION AND OBJECTIVES Vascular endothelial growth factor (VEGF) inhibitors are widely used in oncology and ophthalmology. Although these agents have been shown to increase the risk of cardiovascular events in systemic use, the effect of local applications is unclear. In our study, we aimed to investigate the effects of anti-VEGF agents on left heart functions after intravitreal injection using speckle tracking echocardiography. METHODS In this prospectively designed study, 44 patients who were going to start intravitreal anti-VEGF treatment were included in the study. Patients were evaluated with speckle tracking echocardiography before the first anti-VEGF administration and at three months of anti-VEGF treatment. RESULTS Global longitudinal strain (GLS) values at three months were lower in the patients who participated in the study and this was statistically significant (-18.77±2.17, -18.60±2.01, p=0.001). Also, there was a statistically significant decrease in the mean values of GLS (GLS4CH) obtained from apical four space image, GLS (GLSAPLAX) obtained from apical long axis image and GLS (GLS2CH) obtained from apical 2 space image at month 0 and month 3 (-19.08±2.39, -18.93±2.26, p=0.004; -18.81±2.29, -18.60±2.12, p=0.001; -18.44±2.31, -18.27±2.12, p=0.013, respectively). CONCLUSION The slight decrease in GLS in our study suggests that the use of intravitreal anti-VEGF agents may have cardiac effects.
Collapse
Affiliation(s)
- Mehmet Şahinbaş
- Department of Cardiology, Sivas Numune Hospital, Sivas, Turkey.
| | - İdris Buğra Çerik
- Department of Cardiology, Training and Research Hospital, Ordu University, Ordu, Turkey
| | - Duygu Yalınbaş Yeter
- Department of Ophthalmology, Faculty of Medicine, Cumhuriyet University, Sivas, Turkey
| |
Collapse
|
36
|
Santiago-Sánchez GS, Fabian KP, Hodge JW. A landscape of checkpoint blockade resistance in cancer: underlying mechanisms and current strategies to overcome resistance. Cancer Biol Ther 2024; 25:2308097. [PMID: 38306161 PMCID: PMC10841019 DOI: 10.1080/15384047.2024.2308097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The discovery of immune checkpoints and the development of immune checkpoint inhibitors (ICI) have achieved a durable response in advanced-stage cancer patients. However, there is still a high proportion of patients who do not benefit from ICI therapy due to a lack of response when first treated (primary resistance) or detection of disease progression months after objective response is observed (acquired resistance). Here, we review the current FDA-approved ICI for the treatment of certain solid malignancies, evaluate the contrasting responses to checkpoint blockade in different cancer types, explore the known mechanisms associated with checkpoint blockade resistance (CBR), and assess current strategies in the field that seek to overcome these mechanisms. In order to improve current therapies and develop new ones, the immunotherapy field still has an unmet need in identifying other molecules that act as immune checkpoints, and uncovering other mechanisms that promote CBR.
Collapse
Affiliation(s)
- Ginette S. Santiago-Sánchez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kellsye P. Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
37
|
Fourrier T, Truntzer C, Peroz M, Derangère V, Vincent J, Bengrine-Lefèvre L, Hennequin A, Palmier R, Orry D, Rabel T, Ghiringhelli F. Factors Influencing the Duration of Maintenance Therapy in Metastatic Colorectal Cancer. Cancers (Basel) 2024; 17:88. [PMID: 39796718 PMCID: PMC11720154 DOI: 10.3390/cancers17010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Metastatic colorectal cancer (mCRC) is mainly treated with 5-Fluoro-Uracil (5-FU), Oxaliplatin and Irinotecan chemotherapies and anti-Epidermal Growth Factor Receptor (EGFR) or anti-Vascular Endothelial Growth Factor (VEGF) targeted therapies. Due to chemotherapy-related toxicity, patients receive induction treatment to achieve tumour response followed by maintenance therapy with less cytotoxic molecules or a chemotherapy-free interval to reduce chemotherapy-related toxicity. In this study, the aim was to determine the patient, cancer and treatment factors that influence the duration of maintenance therapy (DMT). METHODS We collected retrospective data on a cohort of 133 patients treated at the Centre Georges François Leclerc (CGFL) cancer centre in Dijon between March 2014 and June 2022. Patients had unresectable or potentially resectable diseases. They received first-line induction treatment with chemotherapy and/or targeted therapy and maintenance treatment, defined as the interruption of at least one chemotherapy agent. RESULTS In the multivariate analysis, age (HR: 1.02, 95% CI 1.00-1.04, p = 0.031), N2 nodal status (HR: 1.78, 95% CI 1.09-2.89, p = 0.021) and the presence of peritoneal metastases (HR: 2.05, 95% CI 1.25-3.36, p = 0.004), as well as baseline carcino-embryonic antigen (CEA) level (HR: 1.10, 95% CI 1.00-1.20, p = 0.052), were significantly associated to poor DMT. Local treatment of liver metastases also significantly reduced the DMT (HR: 0.49, 95% CI 0.28-0.86, p = 0.013). In our cohort, induction triplet chemotherapy significantly increased the CEA delta (70% vs. 44%, p = 0.047) compared to doublet chemotherapy and led to a higher rate of liver surgery (40% vs. 21%, p = 0.014) and a trend for a higher rate of local treatment of metastases (62% vs. 45%, p = 0.059). CONCLUSIONS Duration of maintenance therapy is determined by the initial patient and colorectal cancer characteristics. However, it is significantly increased by local treatment of liver metastases. By reducing the tumour burden, a triplet induction chemotherapy regimen increases the rate of liver metastase resection.
Collapse
Affiliation(s)
- Théo Fourrier
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Caroline Truntzer
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Morgane Peroz
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Valentin Derangère
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| | - Julie Vincent
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Leila Bengrine-Lefèvre
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Rémi Palmier
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - David Orry
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - Thomas Rabel
- Department of Surgical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
| | - François Ghiringhelli
- Cancer Biology Transfer Platform, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- Department of Medical Oncology, Georges François Leclerc Cancer Center, UNICANCER, 21000 Dijon, France
- INSERM UMR1231 Research Center, University of Burgundy, 21000 Dijon, France
| |
Collapse
|
38
|
Sun C, Fan E, Huang L, Zhang Z. Second-line systemic treatment for metastatic colorectal cancer: A systematic review and Bayesian network meta-analysis based on RCT. PLoS One 2024; 19:e0313278. [PMID: 39715232 DOI: 10.1371/journal.pone.0313278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/21/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The optimal second-line systemic treatment for metastatic colorectal cancer (mCRC) is inconclusive. METHODS We searched PubMed, Web of Science, EMBASE, and Cochrane Library for RCTs comparing second-line systemic treatments for mCRC from the inception of each database up to February 3, 2024. Markov Chain Monte Carlo (MCMC) technique was used in this network meta-analysis (NMA) to generate the direct and indirect comparison results among multiple treatments in progression-free survival (PFS), overall response rate (ORR), overall survival (OS), complete response (CR), partial response (PR), grade 3 and above adverse events (Grade ≥ 3AE), and any adverse events (Any AE). The surface under the cumulative ranking curve (SUCRA) was adopted to evaluate the probability of each treatment being the optimum intervention. Subgroup analyses were performed based on the RAS gene status. RESULTS A total of 47 randomized controlled trials were included, involving 16,925 patients and 44 second-line systemic treatments. In improving OS, FOLFOX + Bevacizumab + Erlotinib exhibited significant superiority (SUCRA:92.7%). In improving PFS, Irinotecan + CMAB009 (SUCRA:86.4%) had advantages over other treatments. FOLFIRI + Trebananib (SUCRA:88.1%) had a significant advantage in improving ORR. Among multiple second-line treatments, the SUCRA values of FOLFOX + Bevacizumab in PFS, OS, ORR, and PR were 83.4%, 74.0%, 81.1%, and 86.1%, respectively, and the safety was not significantly different from other interventions. Subgroup analyses showed that FOLFIRI + Bevacizumab + panitumumab ranked among the top in survival outcomes in the RAS-mutant population (OS SUCRA: 87.9%; PFS SUCRA: 70.2%); whereas in the RAS-wild-type population, FOLFIRI + Bevacizumab significantly improved survival outcomes (OS SUCRA: 73.2%; PFS SUCRA: 65.1%). CONCLUSION For most people, FOLFOX + Bevacizumab may be the best second-line systemic treatment regimen for mCRC. For RAS-mutant populations, FOLFIRI + Bevacizumab + Panitumumab is recommended. However, the therapeutic effect may be affected by the patient's physiological state, and clinicians should apply it based on actual conditions.
Collapse
Affiliation(s)
- Chengyu Sun
- Department of Colorectal Surgery, The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Enguo Fan
- State Key Laboratory of Medical Molecular Biology, Department of Microbiology and Parasitology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Luqiao Huang
- Department of Colorectal Surgery, The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| | - Zhengguo Zhang
- Department of Colorectal Surgery, The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou Central Hospital, Xuzhou, Jiangsu, China
| |
Collapse
|
39
|
Ma X, Lu T, Yang Y, Qin D, Tang Z, Cui Y, Wang R. DEAD-box helicase family proteins: emerging targets in digestive system cancers and advances in targeted drug development. J Transl Med 2024; 22:1120. [PMID: 39707322 DOI: 10.1186/s12967-024-05930-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/30/2024] [Indexed: 12/23/2024] Open
Abstract
Cancer has become one of the major diseases threatening human health in the twenty-first century due to its incurability. In 2022, new cases of esophageal and gastrointestinal cancers accounted for 17.1% of all newly diagnosed cancer cases worldwide. Despite significant improvements in early cancer screening, clinical diagnostics, and treatments in recent years, the overall prognosis of digestive system cancer patients remains poor. The DEAD-box helicase family, a crucial member of the RNA helicase family, participates in almost every aspect of RNA metabolism, including transcription, splicing, translation, and degradation, and plays a key role in the occurrence and progression of various cancers. This article aims to summarize and discuss the role and potential clinical applications of DEAD-box helicase family proteins in digestive system cancers. The discussion includes the latest progress in the occurrence, development, and treatment of esophageal and gastrointestinal tumors; the main functions of DEAD-box helicase family proteins; their roles in digestive system cancers, including their relationships with clinical factors; effects on cancer proliferation, migration, and invasion; and involved signaling pathways; as well as the existing inhibitory strategies targeting DDX family proteins, are discussed. Additionally, outlooks on future research directions are provided.
Collapse
Affiliation(s)
- Xiaochao Ma
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Tianyu Lu
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Yue Yang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Da Qin
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Ze Tang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| | - Youbin Cui
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China.
| | - Rui Wang
- Department of Thoracic Surgery, Organ Transplantation Center, the First Hospital of Jilin University, 1 Ximin Street, ChangchunJilin, 130021, China
| |
Collapse
|
40
|
Okawa M, Yamakuchi M, Bibek A, Takenouchi K, Maywar DN, Yamada S, Inoue K, Higurashi K, Nakazawa J, Kawahira M, Kodama T, Tanoue K, Oyama Y, Higashi S, Fujisaki C, Hashinokuchi H, Tabaru A, Kanda H, Tachioka S, Imoto Y, Hashiguchi T, Soga Y. Plasma and serum concentrations of VEGF-A121, but not of VEGF-A165, increase post-bevacizumab administration. PLoS One 2024; 19:e0316035. [PMID: 39700124 DOI: 10.1371/journal.pone.0316035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 12/03/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND VEGF-A concentrations were measured in the blood of bevacizumab-treated cancer patients in previous studies, but a consensus has not formed that would develop VEGF-A into a clinical biomarker. Recently, methods to strictly distinguish between the VEGF-A isoforms have been developed but have not yet been applied to cancer patients undergoing bevacizumab treatment. METHODS An ELISA that strictly distinguishes between VEGF-A121 and VEGF-A165-the major isoforms of VEGF-A-and a commercially available ELISA for VEGF-A are used to determine the concentration of VEGF-A121, VEGF-A165, and VEGF-A in the blood of 12 patients with advanced colorectal cancer receiving bevacizumab therapy. RESULTS The serum and plasma concentrations of VEGF-A121 increased substantially post-bevacizumab administration; the median increase in serum was 860.8 pg/mL, 95% confidence interval (CI) [468.5, 1128.9], p = 0.0024, and in plasma was 808.6 pg/mL, 95% CI [748.7, 874.0], p = 0.00049. In stark contrast, VEGF-A165 after bevacizumab administration decreased in serum by a medium change of -73.8 pg/mL, 95% CI [-149.4, -10.2], p = 0.0034, with 83.3% of the post-bevacizumab concentrations falling below the high-accuracy threshold of 38 pg/mL; in plasma, all pre and post VEGF-A165 concentrations fell below this threshold. Concentrations of VEGF-A121 and VEGF-A165 in platelets did not change to a statistically significant degree. Adding recombinant VEGF-A121 (and -A165) or bevacizumab to plasma in patients post-bevacizumab administration increased or decreased, respectively, VEGF-A121 and VEGF-A165 levels. The increase in VEGF-A121 in plasma and serum after bevacizumab administration may be due to the dissociation of the complex of tumor-derived VEGF-A121 and bevacizumab when it moves from the stroma into the blood. CONCLUSIONS The VEGF-A121 isoform has been uniquely demonstrated as a clear marker of bevacizumab therapy in both plasma and serum, motivating further research on pursuing these isoforms as biomarkers in cancer care.
Collapse
Affiliation(s)
- Masashi Okawa
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Munekazu Yamakuchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Aryal Bibek
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kazunori Takenouchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Drew N Maywar
- Electrical and Computer Engineering Technology, Rochester Institute of Technology, Rochester, New York, United States of America
| | | | | | | | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Masahiro Kawahira
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Tomoko Kodama
- Department of Medical Oncology, Kagoshima City Hospital, Kagoshima, Japan
| | - Kiyonori Tanoue
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Yoko Oyama
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Sadayuki Higashi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Chieko Fujisaki
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | | | - Akito Tabaru
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hideaki Kanda
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shuji Tachioka
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Yutaka Imoto
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Teruto Hashiguchi
- Department of Laboratory and Vascular Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
- Kagoshima University Hospital Clinical Laboratory, Kagoshima, Japan
| | - Yoshiharu Soga
- Department of Cardiovascular and Gastroenterological Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
41
|
Shi M, Yang Y, Huang N, Zeng D, Mo Z, Wang J, Zhang X, Liu R, Wang C, Rong X, Wu Z, Huang Q, Shang H, Tang J, Wang Z, Cai J, Huang G, Guan Y, Guo J, Mu Q, Wang J, Liao W. Genetic and microenvironmental evolution of colorectal liver metastases under chemotherapy. Cell Rep Med 2024; 5:101838. [PMID: 39631402 PMCID: PMC11722126 DOI: 10.1016/j.xcrm.2024.101838] [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: 05/17/2024] [Revised: 09/16/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Drug resistance limits the efficacy of chemotherapy for colorectal cancer liver metastasis (CRLM). However, the evolution of CRLM during drug treatment remains poorly elucidated. Multi-omics and treatment response data from 115 samples of 49 patients with CRLM undergoing bevacizumab (BVZ)-based chemotherapy show little difference in genomic alterations in 92% of cases, while remarkable differences are observed at the transcriptomic level. By decoupling intrinsic and acquired resistance, we find that hepatocyte and myeloid cell infiltration contribute to 38.5% and 23.1% of acquired resistance, respectively. Importantly, SMAD4 mutations and chr20q copy-number gain are associated with intrinsic chemoresistance. Gene interference experiments suggest that SMAD4R361H/C mutations confer BVZ and 5-fluorouracil (5-FU) resistance through STAT3 signaling. Notably, supplementing BVZ and 5-FU with the STAT3 inhibitor GB201 restores therapeutic efficacy in SMAD4R361H/C cancer cells. Our study uncovers the evolutionary dynamics of CRLM and its microenvironment during treatment and offers strategies to overcome drug resistance.
Collapse
Affiliation(s)
- Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Yingxi Yang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Na Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China
| | - Zongchao Mo
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiao Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaomeng Zhang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Ran Liu
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Chunlin Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhenzhen Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Haixia Shang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jihong Tang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Zhaojun Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jianan Cai
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Genjie Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yijin Guan
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian Guo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Quanhua Mu
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Jiguang Wang
- Department of Chemical and Biological Engineering, Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China; SIAT-HKUST Joint Laboratory of Cell Evolution and Digital Health, HKUST Shenzhen-Hong Kong Collaborative Innovation Research Institute, Futian, Shenzhen 518000, P.R. China.
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China; Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China; Foshan Key Laboratory of Translational Medicine in Oncology, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| |
Collapse
|
42
|
Chung SJ, Hadrick K, Nafiujjaman M, Apu EH, Hill ML, Nurunnabi M, Contag CH, Kim T. Targeted Biodegradable Near-Infrared Fluorescent Nanoparticles for Colorectal Cancer Imaging. ACS APPLIED BIO MATERIALS 2024; 7:7861-7870. [PMID: 38574012 PMCID: PMC11653400 DOI: 10.1021/acsabm.4c00072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/06/2024]
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer death in the U.S., and early detection and diagnosis are essential for effective treatment. Current methods are inadequate for rapid detection of early disease, revealing flat lesions, and delineating tumor margins with accuracy and molecular specificity. Fluorescence endoscopy can generate wide field-of-view images enabling detection of CRC lesions and margins; increased signal intensity and improved signal-to-noise ratios can increase both speed and sensitivity of cancer detection. For this purpose, we developed targeted near-infrared (NIR) fluorescent silica nanoparticles (FSNs). We tuned their size to 50-200 nm and conjugated their surface with an antibody to carcinoembryonic antigen (CEA) to prepare CEA-FSNs. The physicochemical properties and biodegradable profiles of CEA-FSN were characterized, and molecular targeting was verified in culture using HT29 (CEA positive) and HCT116 (CEA negative) cells. CEA-FSNs bound to the HT29 cells to a greater extent than to the HCT116 cells, and smaller CEA-FSNs were internalized into HT29 cells more efficiently than larger CEA-FSNs. After intravenous administration of CEA-FSNs, a significantly greater signal was observed from the CEA-positive HT29 than the CEA-negative HCT116 tumors in xenografted mice. In F344-PIRC rats, polyps in the intestine were detected by white-light endoscopy, and NIR fluorescent signals were found in the excised intestinal tissue after topical application of CEA-FSNs. Immunofluorescence imaging of excised tissue sections demonstrated that the particle signals coregistered with signals for both CRC and CEA. These results indicate that CEA-FSNs have potential as a molecular imaging marker for early diagnosis of CRC.
Collapse
Affiliation(s)
- Seock-Jin Chung
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kay Hadrick
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Md Nafiujjaman
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Ehsanul Hoque Apu
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Meghan L. Hill
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Md Nurunnabi
- Department
of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas 79902, United States
| | - Christopher H. Contag
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Microbiology, Genetics and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Taeho Kim
- Department
of Biomedical Engineering, Institute for Quantitative Health Science
and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
43
|
Hsiao KY, Chen HP, Rau KM, Liu KW, Shia BC, Chang WS, Liang HY, Hsieh MC. Association between sidedness and survival among chemotherapy refractory metastatic colorectal cancer patients treated with trifluridine/tipiracil or regorafenib. Oncologist 2024; 29:e1669-e1679. [PMID: 39245044 PMCID: PMC11630785 DOI: 10.1093/oncolo/oyae235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 08/05/2024] [Indexed: 09/10/2024] Open
Abstract
BACKGROUND The impact of sidedness on survival of later-line treatment in patients with metastatic colorectal cancer (mCRC) is undetermined. This study aimed to investigate the association between sidedness and survival among chemotherapy refractory patients with mCRC treated with trifluridine/tipiracil (TAS-102) or regorafenib or both. PATIENTS AND METHODS Patients with mCRC treated with TAS-102 or regorafenib between 2015 and 2020 was retrospectively collected. Patients were stratified into TAS-102 first and regorafenib first, then subdivided into TAS-102 followed by regorafenib (T-R) and regorafenib followed by TAS-102 (R-T) groups. The oncologic outcomes were presented with time-to-treatment failure (TTF) and overall survival (OS). RESULTS After matching, 376 TAS-102 patients and 376 regorafenib patients were included for outcomes comparison. TTF had insignificant differences while OS was significantly different between TAS-102 and regorafenib groups. Median TTF and OS were 1.9 months versus 2.0 months (P = .701) and 9.1 months versus 7.0 months (P = .008) in TAS-102 and regorafenib, respectively. The OS benefits were consistent regardless primary tumor location. Subgroup analysis with 174 T-R patients and 174 R-T patients was investigated for treatment sequences. TTF and OS had significant differences in both groups. Median TTF and OS were 8.5 months versus 6.3 months (P = .001) and 14.4 months versus 12.6 months (P = .035) in T-R and R-T groups, respectively. The TTF and OS benefits were persisted regardless primary tumor location. CONCLUSION TAS-102 first provided a better survival benefit in chemotherapy refractory patients with mCRC across all sidedness. Further prospective studies are warranted to validate our conclusions.
Collapse
Affiliation(s)
- Kai-Yuan Hsiao
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hsin-Pao Chen
- Division of Colon and Rectum Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, Taiwan
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Kun-Ming Rau
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung, Taiwan
| | - Kuang-Wen Liu
- Division of Colon and Rectum Surgery, Department of Surgery, E-Da Hospital, Kaohsiung, Taiwan
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Ben-Chang Shia
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wei-Shan Chang
- Artificial Intelligence Development Center, Fu Jen Catholic University, New Taipei City, Taiwan
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Hao-Yun Liang
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- EMMT Systems Corporation, Director, Taiwan
| | - Meng-Che Hsieh
- College of Medicine, I-Shou University, Kaohsiung, Taiwan
- Department of Hematology-Oncology, E-Da Cancer Hospital, Kaohsiung, Taiwan
| |
Collapse
|
44
|
Banerjee N, Roy L, Panda S, Roychowdhury T, Chatterjee S. In Silico-Designed G-Quadruplex Targeting Peptide Attenuates VEGF-A Expression, Preventing Angiogenesis in Cancer Cells. Chem Biol Drug Des 2024; 104:e70018. [PMID: 39704035 DOI: 10.1111/cbdd.70018] [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: 05/28/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 12/21/2024]
Abstract
Vascular endothelial growth factor-A (VEGF-A) is a growth factor and pluripotent cytokine that promotes angiogenesis in cancer cells, transitioning to an angiogenic phenotype. The binding of VEGF-A protein to VEGF receptors (VEGFR-1 and VEGFR-2) initiates a cascade of events that stimulates angiogenesis by facilitating the migration and enhancing the permeability of endothelial cells. The proximal promoter of the VEGF gene encompasses a 36-base pair region (from -85 to -50) that can form a stable G-quadruplex (G4) structure in specific conditions. The activity of the VEGF promoter is reliant on this structure. During cancer progression, the VEGF-A G4 succumbs to cellular pressure and fails to maintain a stable structure. This shifts the balance to form a duplex structure, increasing the transcription rate. Earlier research has tried to develop small-molecule ligands to target and stabilise G4, demonstrating the possibility of suppressing VEGF expression. However, they either lack specificity or toxic. Peptides, on the other hand, are significantly less studied as G4 binders. Here, we designed a peptide that successfully binds and stabilises the VEGF-A G4 while reducing its gene expression. This further alters the expression fate of the VEGF-A signalling cascade and blocks angiogenesis in cancer cells. We employed high-resolution nuclear magnetic resonance (NMR) spectroscopy and molecular dynamics simulation to elucidate the chemical details of G4-peptide interaction. In addition, we used qPCR and western blot techniques to investigate the expression pattern of the molecules implicated in the VEGF-A signalling cascade. The study explores the intricate relationship between peptides and quadruplex structures, revealing valuable insights that can improve the design of pharmacophores targeting the dynamic quadruplex structure. The results of our study are encouraging, opening possibilities for advancements in, the characterisation and optimisation of peptides as G-quadruplex ligands in view of their potential therapeutic uses.
Collapse
Affiliation(s)
- Nilanjan Banerjee
- Non-Coding Genome Group, Department of Structural Biology, CEITEC-Central European Institute of Technology, Brno, Czech Republic
| | - Laboni Roy
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| | - Suman Panda
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| | - Tanaya Roychowdhury
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Subhrangsu Chatterjee
- Department of Biological Science, Bose Institute, Unified Academic Campus, Kolkata, India
| |
Collapse
|
45
|
Roazzi L, Patelli G, Bencardino KB, Amatu A, Bonazzina E, Tosi F, Amoruso B, Bombelli A, Mariano S, Stabile S, Porta C, Siena S, Sartore-Bianchi A. Ongoing Clinical Trials and Future Research Scenarios of Circulating Tumor DNA for the Treatment of Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:295-308. [PMID: 38519391 DOI: 10.1016/j.clcc.2024.02.001] [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: 09/14/2023] [Revised: 01/04/2024] [Accepted: 02/11/2024] [Indexed: 03/24/2024]
Abstract
Liquid biopsy using circulating tumor DNA (ctDNA) has emerged as a minimally invasive, timely approach to provide molecular diagnosis and monitor tumor evolution in patients with cancer. Since the molecular landscape of metastatic colorectal cancer (mCRC) is substantially heterogeneous and dynamic over space and time, ctDNA holds significant advantages as a biomarker for this disease. Numerous studies have demonstrated that ctDNA broadly recapitulates the molecular profile of the primary tumor and metastases, and have mainly focused on the genotyping of RAS and BRAF, that is propaedeutic for anti-EGFR treatment selection. However, ctDNA soon broadened its scope towards the assessment of early tumor response, as well as the identification of drug resistance biomarkers to drive potential molecular actionability. In this review article, we provide an overview of the current state-of-the-art of this methodology and its applications, focusing on ongoing clinical trials that employ ctDNA to prospectively guide treatment in patients with mCRC.
Collapse
Affiliation(s)
- Laura Roazzi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Katia Bruna Bencardino
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alessio Amatu
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Tosi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Brunella Amoruso
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Bombelli
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sara Mariano
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Stefano Stabile
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, A.O.U. Consorziale Policlinico di Bari, Bari, Italy; Interdisciplinary Department of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy; Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy.
| |
Collapse
|
46
|
Lu B, Dvorani E, Nguyen L, Beca JM, Mercer RE, Adamic A, Muñoz C, Chan KKW. Cost-Effectiveness Analysis of Bevacizumab Biosimilars Versus Originator Bevacizumab for Metastatic Colorectal Cancer: A Comparative Study Using Real-World Data. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2024; 27:1689-1697. [PMID: 39127249 DOI: 10.1016/j.jval.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 06/10/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVES MVASI (Amgen) and Zirabev (Pfizer) are 2 of the earliest bevacizumab biosimilars approved for the first-line treatment of metastatic colorectal cancer (mCRC). We aimed to confirm and quantify the real-world cost savings and cost-effectiveness of MVASI and Zirabev relative to originator bevacizumab for patients with mCRC. METHODS We conducted a population-based, retrospective cohort study in Ontario, Canada, where originator and biosimilar bevacizumab are universally publicly funded. All mCRC patients who received originator bevacizumab between January 2008 and August 2019 or biosimilar bevacizumab between August 2019 and March 2021 were propensity score matched (1:4) to adjust for baseline differences. Total 1-year patient-level costs (CAD) and effects (life years [LY] and quality-adjusted LYs) were calculated from the public health payer's perspective. Primary outcomes included incremental net monetary benefit and incremental net health benefit (INHB). Sensitivity analyses included a subgroup analysis by biosimilar type (MVASI/Zirabev) and a 2-year analysis. RESULTS The matched cohort included 747 biosimilar cases and 2945 comparators. Bevacizumab biosimilars were associated with an incremental cost of -$6379 (95%CI: -9417, -3537) (ie, cost saving) and incremental effect of 0.0 (95% CI: -0.02, 0.02) LY and -0.01 (95% CI: -0.03, 0) quality-adjusted LYs gained. Incremental net monetary benefit and INHB estimates were $6331 (95% CI: 6245, 6417) and 0.127 LY (95% CI: 0.125, 0.128), respectively, at a willingness-to-pay threshold of $50 000/life year gained, with all estimates indicating the cost-effectiveness of biosimilar bevacizumab. Cost-effectiveness remained consistent across biosimilar brand subgroups and 2-year sensitivity analyses. CONCLUSION Bevacizumab biosimilars demonstrated real-world cost savings while providing similar survival benefit as originator bevacizumab, confirming the initial expectations of their implementation and supporting health system sustainability.
Collapse
Affiliation(s)
- Brandon Lu
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | | | - Jaclyn M Beca
- Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada; Morse Consulting Inc., Toronto, ON, Canada
| | - Rebecca E Mercer
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada; Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
| | - Andrea Adamic
- Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada; Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
| | - Caroline Muñoz
- Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada; Ontario Health (Cancer Care Ontario), Toronto, ON, Canada
| | - Kelvin K W Chan
- Sunnybrook Health Sciences Centre, Toronto, ON, Canada; ICES, Toronto, ON, Canada; Canadian Centre for Applied Research in Cancer Control, Toronto, ON, Canada; Morse Consulting Inc., Toronto, ON, Canada.
| |
Collapse
|
47
|
Sroor FM, El-Sayed AF, Mahmoud K. Novel 5-Fluorouracil analogues versus perfluorophenyl ureas as potent anti-breast cancer agents: Design, robust synthesis, in vitro, molecular docking, pharmacokinetics ADMET analysis and dynamic simulations. Bioorg Chem 2024; 153:107944. [PMID: 39532011 DOI: 10.1016/j.bioorg.2024.107944] [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: 09/22/2024] [Revised: 10/22/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
To investigate the therapeutic potential of 5-Fluorouracil-based analogues, a straightforward synthetic technique was employed to synthesize a novel series of 5-arylurea uracil derivatives (AUFU01-03) and aryl-urea derivatives bearing perfluorophenyl (AUPF01-03). Reliable tools such as infrared (IR), Nuclear Magnetic Resonance (NMR) spectra, and elemental analyses were utilized to confirm the chemical structures and purity of these compounds. In comparison to healthy noncancerous control skin fibroblast cells (BJ-1), we examined the antiproliferative efficacy of compounds (AUFU01-03) and (AUPF01-03) against specific human malignant cell lines of the breast (MCF-7), and colon (HCT-116). Based on the MTT experiment results, compounds AUFU03 and AUPF01-03 possessed highly cytotoxic effects. Among these, cytotoxicity was demonstrated by compounds AUPF01-03 with IC50 values (AUPF01, IC50 = 167 ± 0.57 µM, AUPF02, IC50 = 23.4 ± 0.68 µM and AUPF03, IC50 = 28.8 ± 1.13 µM, respectively, on MCF-7), relative to 5-Fluorouracil as reference drug (IC50 = 160.7 ± 0.22 µM). Compound AUPF01 showed safety on BJ-1 cells up to a concentration of 100 µM (% cytotoxicity = 3.9 ± 0.42 %), so AUPF01 was selected for further studies. At the gene, the expression levels of BCL-2 gene were decreased significantly in MCF-7 + 5-FU and reached the lowest level in MCF-7 + AUPF01. In contrast, the expression levels of pro-apoptotic genes (p53 and BAX) were increased in MCF-7 + 5-FU, and reached a significantly higher level in MCF-7 + AUPF01. Apoptosis/necrosis assays demonstrated that AUPF01 induced S and G2/M phase cell cycle arrest in MCF-7 cells. Moreover, the efficacy of these compounds against anti-cancer protein receptors was assessed using molecular docking. The results indicated that compound AUPF01 exhibited high binding energies, effectively interacting with the active sites of crucial proteins such as EGFR, CDK2, ERalfa, BAX1, BCL2, and P53. These interactions involved a diverse range of chemical bonding types, suggesting the potential of these substances to inhibit enzyme activities. Moreover, computational ADMET analyses of these compounds demonstrated compliance with Lipinski's criteria, indicating favorable physicochemical properties. Additionally, molecular dynamics (MD) simulations revealed stable complexes of AUPF01 with EGFR, CDK2, ERalfa, BAX1, BCL2, and P53, as evidenced by (RMSD) values, RMSF values, and (SASA) values for the respective complexes.
Collapse
Affiliation(s)
- Farid M Sroor
- Organometallic and Organometalloid Chemistry Department, National Research Centre, 12622 Cairo, Egypt.
| | - Ahmed F El-Sayed
- Microbial Genetics Department, Biotechnology Research Institute, National Research Centre, Giza, Egypt; Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo, Egypt
| | - Khaled Mahmoud
- Pharmacognosy Department, Pharmaceutical and Drug Industry Institute, National Research Centre, 12622 Dokki, Egypt
| |
Collapse
|
48
|
Samanta S, Sarkar S, Kayal K. Cancer model and its possible control-A Z-type control approach. MethodsX 2024; 13:102895. [PMID: 39687596 PMCID: PMC11647952 DOI: 10.1016/j.mex.2024.102895] [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: 05/16/2024] [Revised: 07/28/2024] [Accepted: 08/06/2024] [Indexed: 12/18/2024] Open
Abstract
This paper investigates the dynamics of a three-dimensional nonlinear cancer model involving interactions among cancer cells, normal cells, and immune cells. By performing a linear stability analysis of the equilibria and investigating the Hopf bifurcation in relation to the immune cell growth rate, we reveal the possibility of chaotic behavior when radiation is absent. However, with the appropriate implementation of radiotherapy, the cancer model demonstrates stable solutions, transitioning from chaotic oscillations through period-halving bifurcation. Additionally, we propose and examine an indirect Z-control mechanism within the cancer model. Our findings indicate that using the indirect Z-controller on the immune population successfully manages chaos and adjusts the cancer cell density to a desired level. Through extensive investigation, we demonstrate the robustness of the Z-controller in managing oscillations and provide insights into determining the minimum number of immune cells needed to achieve a predetermined cancer cell density. This study underscores the importance of control mechanisms in mitigating cancer progression and highlights the potential of Z-control for therapeutic intervention strategies.
Collapse
Affiliation(s)
- Sudip Samanta
- Department of Mathematics, Bankura University, Bankura 722155, India
| | - Sandip Sarkar
- Sri Aurobindo Vidyamandir, Chandannagar, Hooghly, West Bengal 712136, India
| | - Kaushik Kayal
- Agricultural and Ecological Research Unit, Indian Statistical Institute, 203, B. T. Road, Kolkata 700108, India
| |
Collapse
|
49
|
Luengas‐Martinez A, Ismail D, Paus R, Young HS. Vascular endothelial growth factor A inhibition remodels the transcriptional signature of lipid metabolism in psoriasis non-lesional skin in 12 h ex vivo culture. SKIN HEALTH AND DISEASE 2024; 4:e471. [PMID: 39624732 PMCID: PMC11608907 DOI: 10.1002/ski2.471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Vascular endothelial growth factor A (VEGF-A)-mediated angiogenesis is involved in the pathogenesis of psoriasis. VEGF-A inhibitors are widely used to treat oncological and ophthalmological diseases but have not been used in psoriasis management. The molecular mechanisms underlying the effects of VEGF-A inhibition in psoriatic skin remain unknown. OBJECTIVES To identify the genes and canonical pathways affected by VEGF-A inhibition in non-lesional and plaque skin ex vivo. METHODS Total RNA sequencing was performed on skin biopsies from patients with psoriasis (n = 6; plaque and non-lesional skin) and healthy controls (n = 6) incubated with anti-VEGF-A monoclonal antibody (bevacizumab, Avastin®) or human IgG1 isotype control for 12 h in serum-free organ culture. Differentially expressed genes between paired control and treated samples with adjusted p-values <0.1 were considered significant. Gene ontology and ingenuity pathway analysis was used to identify enriched biological processes, canonical pathways and upstream regulators. RESULTS VEGF-A inhibition upregulated the expression of genes involved in lipid metabolism. Pathway enrichment analysis identified the activation of pathways involved in fatty acids and lipid biosynthesis and degradation in non-lesional skin and ferroptosis in plaque skin. VEGF-A inhibition downregulated endothelial cell apoptosis in non-lesional psoriasis skin and members of the interferon family were identified as potential regulators of the effects of VEGF-A inhibition in non-lesional skin. CONCLUSION Early response to VEGF-A inhibition is associated with changes in lipid metabolism in non-lesional psoriasis skin and cellular stress in psoriasis plaque. More investigation is needed to validate these findings.
Collapse
Affiliation(s)
- Andrea Luengas‐Martinez
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Dina Ismail
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| | - Ralf Paus
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
- Dr. Philip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Monasterium LaboratoryMuensterGermany
| | - Helen S. Young
- Centre for Dermatology Research and Manchester Academic Health Science CentreThe University of ManchesterManchesterUK
| |
Collapse
|
50
|
Mo C, Chadha B, Kuang C. An Evolving Landscape: New Therapies for Metastatic Colorectal Cancer. Clin Colorectal Cancer 2024; 23:337-345. [PMID: 39332920 PMCID: PMC11608151 DOI: 10.1016/j.clcc.2024.08.003] [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: 07/20/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/29/2024]
Abstract
Substantial progress is being made in the development of novel therapies directed against colorectal cancer. The discovery of various molecular markers and advances in tumor profiling have facilitated the development of new targeted agents and immunotherapy. Not only have these drugs improved progression-free survival and even overall survival in some cases, but their related outcomes have also raised questions as to how to best combine or sequence therapies for even greater efficacy. Furthermore, we are beginning to understand how these combination therapies may yield for greater therapeutic response for patients with microsatellite stable colorectal cancer for which there is much need for improvement. In this article, we review recent trial data and explore the outcomes of various targeted therapies and immunotherapies for patient with advanced colorectal cancer.
Collapse
Affiliation(s)
- Christiana Mo
- Department of Oncology, Montefiore Einstein, Bronx, NY; Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
| | - Bhawneet Chadha
- Department of Oncology, Montefiore Einstein, Bronx, NY; Montefiore Einstein Comprehensive Cancer Center, Bronx, NY
| | - Chaoyuan Kuang
- Department of Oncology, Montefiore Einstein, Bronx, NY; Department of Molecular Pharmacology, Montefiore Einstein, Bronx, NY.
| |
Collapse
|