1
|
Verschuere H, Kasmi S, Nuhn L, D'Almeida SM, Zhu Q, Zhong Z, Adjemian S, Louage B, De Vrieze J, Yu H, De Geest BG, Vandenabeele P. Enhancing anti-tumor immunity through intratumoral combination therapy with amphiphilic conjugates of oxaliplatin and imidazoquinoline TLR7/8 agonist. RSC Adv 2025; 15:11662-11674. [PMID: 40230629 PMCID: PMC11995270 DOI: 10.1039/d5ra00163c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
The efficacy of conventional chemotherapy does not only rely on the cytotoxic action of the drug compound itself. Indeed, proper drug-induced immunogenic cell death (ICD) can stimulate immunosurveillance and mount a systemic anti-tumor response. We aimed to further amplify the therapeutic activity of oxaliplatin (OxPt) chemotherapy-induced ICD by combining this with an imidazoquinoline (IMDQ) TLR7/8 agonist. We hypothesized that innate immune activation by TLR7/8 activation primes the immune system against tumor neoantigens, thereby mounting tumor-specific T cell responses that contribute to killing primary tumor cells and distal metastases. To this end, we initially synthesized a covalent conjugate of OxPt, an imidazoquinoline TLR7/8 agonist (i.e., IMDQ), and an alkyl lipid. We hypothesized that such a lipidated conjugate would, upon intratumoral injection, increase the residence time in the tumor and reduce systemic dissemination and, hence, off-target toxicity. Whereas combination therapy with OxPt and IMDQ in native form improved, relative to single treatment, the anti-tumor efficacy against the primary treated tumor and a secondary distal tumor, this was not the case for OxPt-IMDQ-lipid conjugate therapy. We then altered the molecular design of the combination therapy and synthesized amphiphilic OxPt and IMDQ conjugates, comprising a cholesteryl motif and a hydrophilic poly(ethylene glycol) (PEG) chain. Intratumoral combination therapy with OxPt-PEG-cholesteryl and IMDQ-PEG-cholesteryl reduced, compared to native drug compounds, systemic innate inflammatory responses, and more efficiently eradicated primary and distal tumors. Furthermore, we found that combination therapy with OxPt-PEG-cholesteryl and IMDQ-PEG-cholesteryl induced antigen-specific anti-tumor responses and high infiltration levels of CD8+ T cells into the tumor.
Collapse
Affiliation(s)
- Hanne Verschuere
- Cell Death and Inflammation Unit, VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
| | - Sabah Kasmi
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Department of Pharmaceutics, Ghent University Ghent Belgium
| | - Lutz Nuhn
- Department of Chemistry and Pharmacy, Julius-Maximilians-Universität Würzburg Würzburg Germany
| | - Sènan Mickaël D'Almeida
- CyTOF Flow Cytometry Core Facility, École Polytechnique Fédérale de Lausanne (EPFL) Lausanne Switzerland
| | - Qiwen Zhu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Zifu Zhong
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Department of Pharmaceutics, Ghent University Ghent Belgium
| | - Sandy Adjemian
- Cell Death and Inflammation Unit, VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
| | - Benoit Louage
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Department of Pharmaceutics, Ghent University Ghent Belgium
| | - Jana De Vrieze
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Department of Pharmaceutics, Ghent University Ghent Belgium
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences Shanghai 201203 China
| | - Bruno G De Geest
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Department of Pharmaceutics, Ghent University Ghent Belgium
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University Belgium
- Methusalem Program, Ghent University Belgium
| |
Collapse
|
2
|
Fauvre A, Ursino C, Garambois V, Culerier E, Milazzo LA, Vezzio-Vié N, Jeanson L, Marchive C, Andrade AF, Combes E, Atis S, Lossaint G, Quenet F, Michaud HA, Khellaf L, Corbeau I, Tosi D, Houede N, Bonnefoy N, Sgarbura O, Gongora C, Faget J. Oxaliplatin, ATR inhibitor and anti-PD-1 antibody combination therapy controls colon carcinoma growth, induces local and systemic changes in the immune compartment, and protects against tumor rechallenge in mice. J Immunother Cancer 2025; 13:e010791. [PMID: 40139833 PMCID: PMC11950992 DOI: 10.1136/jitc-2024-010791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common cancer type and one of the leading causes of cancer-related death worldwide. The treatment of advanced metastatic CRC relies on classical chemotherapy combinations (5-fluorouracil, oxaliplatin or irinotecan). However, their use is limited by the emergence of resistance mechanisms, including to oxaliplatin. In this context, we recently showed that the combination of oxaliplatin and ataxia telangiectasia and Rad3-related protein inhibition (VE-822) is synergistic and may have a potential therapeutic effect in metastatic CRC management. METHODS In this study, we investigated the role of the VE-822+oxaliplatin (Vox) combination on the immune response and its potential synergy with an anti-programmed-cell Death receptor-1 (PD-1) antibody. We used cell lines and organoids from metastatic CRC to investigate in vitro Vox efficacy and orthotopic syngeneic mouse models of metastatic CRC to assess the efficacy of Vox+anti-PD-1 antibody and identify the involved immune cells. RESULTS The Vox+anti-PD-1 antibody combination completely cured tumor-bearing mice and protected them from a rechallenge. Vox was associated with a reduction of tumor-infiltrated neutrophils, CD206+ macrophages and regulatory T cells. Vox also induced a deep depletion of blood neutrophils. The increased bone marrow granulopoiesis failed to compensate for the Vox-mediated mature neutrophil depletion. Neutrophil depletion using a mouse recombinant anti-Ly6G antibody partially mimicked the Vox effect on the tumor microenvironment, but to a lower extent compared with the Vox+anti-PD-1 antibody combination. Vox, but not neutrophil depletion, led to the emergence of an Ly6C+ PD-1+ CD8+ T-cell population in the blood and spleen of tumor-harboring mice. These cells were proliferating, and expressed IFN-γ, CD62L, CXCR3 and Eomes. Moreover, the proportion of tumor antigen-specific T cells and of CD122+ BCL6+ T cells, which shared phenotypic characteristics with stem-like CD8+ T cells, was increased in treated mice. CONCLUSIONS Our work strongly suggests that the Vox+anti-PD-1 antibody combination might significantly improve survival in patients with metastatic and treatment-refractory CRC by acting both on cancer cells and CD8+ T cells.
Collapse
Affiliation(s)
- Alexandra Fauvre
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Chiara Ursino
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Veronique Garambois
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Elodie Culerier
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Louis-Antoine Milazzo
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Nadia Vezzio-Vié
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Laura Jeanson
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Candice Marchive
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Augusto Faria Andrade
- McGill University/Research Institute of McGill University, Nada Jabado Lab, Montreal, Quebec, Canada
| | - Eve Combes
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Salima Atis
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Gérald Lossaint
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - François Quenet
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Henri-Alexandre Michaud
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Lakhdar Khellaf
- Department of Pathology, Montpellier University, Montpellier, France
| | - Ileana Corbeau
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Diego Tosi
- Medical Oncology Department, Institut régional du Cancer de Montpellier, Montpellier, France
| | - Nadine Houede
- Department of Oncology, University Hospital of Nimes, Nîmes, France
| | - Nathalie Bonnefoy
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Olivia Sgarbura
- Institut regional du Cancer de Montpellier, Montpellier, France
| | - Céline Gongora
- Résistance aux traitements et thérapies innovantes, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), CNRS, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
- CNRS, Paris, France
| | - Julien Faget
- Immunity and Cancer Team, Institut de Recherche en Cancérologie de Montpellier (IRCM), Université de Montpellier (UM), Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
3
|
Wang M, Wang X. Chemoprotective Potential of Cyanidin-3-Glucoside Against 1,2-Dimethylhydrazine-Induced Colorectal Cancer: Modulation of NF-κB and Bcl-2/Bax/Caspase Pathway. J Biochem Mol Toxicol 2025; 39:e70125. [PMID: 39843995 DOI: 10.1002/jbt.70125] [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: 11/03/2024] [Revised: 11/30/2024] [Accepted: 12/21/2024] [Indexed: 01/24/2025]
Abstract
Colorectal cancer (CRC) represents a significant global health challenge, with approximately 1.8 million new cases diagnosed annually and a mortality toll exceeding 881,000 lives each year. This study aimed to evaluate the chemoprotective efficacy of Cyanidin-3-glucoside (C3G) in a rat model of CRC induced by 1,2-dimethylhydrazine (DMH). Rats were stratified into groups and administered C3G at doses of 10 and 15 mg/kg following DMH exposure to initiate CRC. Key parameters, including organ weights, tumor burdens, and biochemical markers, were meticulously assessed. Administration of C3G significantly restored body weight while reducing the weights of colon and spleen tissues. Moreover, C3G treatment substantially suppressed tumor incidence and weight in DMH-induced CRC rats. Biochemical analysis revealed that C3G markedly reduced levels of CFA, CA19.9, LDH, and nitric oxide (NO). It also modulated lipid profiles, antioxidant activities, and the expression of both Phase I and II enzymes. Inflammatory mediators, including TNF-α, IL-1β, IL-1α, IL-2, IL-4, IL-6, IL-10, IL-12, and IL-17, were significantly downregulated. Notably, C3G inhibited inflammatory markers such as COX-2, PGE2, iNOS, and NF-κB while promoting Caspase-3, -6, and -9 activity. Furthermore, it regulated the Bax/Bcl-2 apoptotic axis, reducing the Bcl-2/Bax ratio. Cyanidin-3-glucoside demonstrated potent chemopreventive effects against colorectal cancer in this experimental model. Its mechanism of action is likely mediated through modulation of NF-κB and the Bcl-2/Bax/Caspase pathway, suggesting its potential as a therapeutic agent in CRC management.
Collapse
Affiliation(s)
- Miao Wang
- Department of Gastroenterology, The Second Hospital of Heilongjiang Province, Harbin City, Heilongjiang Province, China
| | - Xiaoyong Wang
- Department of Gastroenterology, The Second Hospital of Heilongjiang Province, Harbin City, Heilongjiang Province, China
| |
Collapse
|
4
|
Rudalska R, Harbig J, Forster M, Woelffing P, Esposito A, Kudolo M, Botezatu A, Haller V, Janssen N, Holzmayer S, Nahidino P, Trompak O, Pantsar T, Kronenberger T, Yurttas C, Rist E, Weber ANR, Dahlke MH, Ott G, Koenigsrainer A, Rothbauer U, Maerklin M, Muerdter T, Schwab M, Singer S, Zender L, Laufer S, Dauch D. First-in-class ultralong-target-residence-time p38α inhibitors as a mitosis-targeted therapy for colorectal cancer. NATURE CANCER 2025; 6:259-277. [PMID: 39820127 PMCID: PMC11864979 DOI: 10.1038/s43018-024-00899-7] [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: 04/19/2023] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Colorectal cancer (CRC) constitutes the second leading cause of cancer-related death worldwide and advanced CRCs are resistant to targeted therapies, chemotherapies and immunotherapies. p38α (Mapk14) has been suggested as a therapeutic target in CRC; however, available p38α inhibitors only allow for insufficient target inhibition. Here we describe a unique class of p38α inhibitors with ultralong target residence times (designated ULTR-p38i) that robustly inhibit p38α downstream signaling and induce distinct biological phenotypes. ULTR-p38i monotherapy triggers an uncontrolled mitotic entry by activating Cdc25 and simultaneously blocking Wee1. Consequently, CRC cells undergo mitotic catastrophe, resulting in apoptosis or senescence. ULTR-p38i exhibit high selectivity, good pharmaco-kinetic properties and no measurable toxicity with strong therapeutic effects in patient-derived CRC organoids and syngeneic CRC mouse models. Conceptually, our study suggests ultralong-target-residence-time kinase inhibitors as an alternative to covalent inhibitors, which, because of the lack of cysteine residues, cannot be generated for many kinase cancer targets.
Collapse
Affiliation(s)
- Ramona Rudalska
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Jule Harbig
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Michael Forster
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Pascal Woelffing
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Aylin Esposito
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Mark Kudolo
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Adelina Botezatu
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Vanessa Haller
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Samuel Holzmayer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Philipp Nahidino
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Omelyan Trompak
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Tatu Pantsar
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Thales Kronenberger
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Elke Rist
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Marc H Dahlke
- Department of General and Visceral Surgery, Robert Bosch Hospital, Stuttgart, Germany
| | - German Ott
- Department of Clinical Pathology, Robert Bosch Hospital, Stuttgart, Germany
| | - Alfred Koenigsrainer
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department of Pharmaceutical Biotechnology, University of Tübingen, Tübingen, Germany
| | - Melanie Maerklin
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Muerdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Singer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Stefan Laufer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Daniel Dauch
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany.
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany.
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany.
| |
Collapse
|
5
|
Cheng Y, Luo R, Li E. Combined delivery of IL12 and an IL18 mutant without IL18BP-binding activity by an adenoviral vector enhances tumor specific immunity. Sci Rep 2025; 15:3563. [PMID: 39875404 PMCID: PMC11775126 DOI: 10.1038/s41598-024-84693-6] [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: 08/13/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
Cytokines play pivotal roles in anticancer immune response. We previously reported that adenovirus armed with an IL18 variant (DR18) that overcomes IL18BP neutralizing effect displayed powerful therapeutic effects in local and distant tumors when delivered intratumorally. Here, we tested a combined delivery of IL12 and DR18 in tumor models since IL12 and IL18 are known to act synergistically in potentiating IFNγ production and antitumor immunity. To minimize adverse effects associated with systemic delivery, we constructed oncolytic adenoviruses (oAd) harboring DR18 and IL12 (oAd.DR18/IL12). IL12 was expressed as a single chain IL12 (scIL12) peptide composed of the IL12/p40 and IL12/p35 subunits. Intratumoral administration of oAd.DR18/IL12, oAd-expressing DR18 (oAd.DR18), or oAd-expressing IL12 (oAd.IL12) showed antitumor effect in syngeneic colorectal tumor models. Compared to oAd.DR18 or oAd.IL12, administration of oAd.DR18/IL12 improved the antitumor effects as well as increased survival rate in these models. We detected enhanced tumor infiltrating T lymphocytes and NK cells in oAd.DR18/IL12-treated mice than those from mock-treated or individually treated groups. Moreover, mice received oAd.DR18/IL12 had more robust tumor-specific cytotoxicity. Importantly, mice that had tumor regression after oAd.DR18/IL12 treatment established anti-tumor specific immune memory. These results show that adenovirus armed with engineered cytokines boosts tumor specific immunity and antitumor effect.
Collapse
Affiliation(s)
- Yan Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Department of Blood Screening Laboratory, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Renjie Luo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Erguang Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.
- Institute of Medical Virology, Medical School, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Warwar SC, Janczewski LM, Rodriguez GM, Wayne JD, Bentrem DJ. Trends in Immunotherapy (IO) Use and Survival Among Patients With High-Incidence Stage IV Cancers Across the United States. J Surg Oncol 2025. [PMID: 39806540 DOI: 10.1002/jso.28084] [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: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND AND OBJECTIVES IO has transformed cancer management, but its adoption in advanced cancer patients varies by tumor type. With more Stage IV patients undergoing surgery, understanding site-specific outcomes in these challenging patients is essential. We aimed to evaluate IO use and survival trends for Stage IV cancer patients across high-incidence cancers in the US. METHODS Patients diagnosed with Stage IV prostate, breast, melanoma, colorectal, renal, bladder, lung, or pancreas cancer were identified from the National Cancer Database (2004-2020). Cochrane-Armitage test and Kaplan-Meier methods assessed IO and overall survival across three periods: 2004-2010, 2011-2015, and 2016-2020. RESULTS Among 1 425 731 Stage IV cancer patients, most had lung (50.0%), pancreas (12.5%), and breast cancer (9.3%), while the least had melanoma (2.2%). From periods 1 to 3, IO use increased from 1.0% to 24.6%, notably in melanoma (9.5% to 58.5%, p < 0.001). Melanoma exhibited the greatest survival gains (median survival: 7.1 to 14.9 months). Absolute increases in 3-year overall survival rates ranged from 3.4% in pancreas (1.7% to 5.1%) to 21.4% in melanoma (15.7% to 37.1%). CONCLUSIONS Utilization of IO is tumor-site specific and associated with improved survival rates for Stage IV cancer, with varied success across types. Variations in receipt highlight ongoing challenges to ensure equitable adoption.
Collapse
Affiliation(s)
- Samantha C Warwar
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lauren M Janczewski
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Gladys M Rodriguez
- Division of Hematology and Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Jeffrey D Wayne
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - David J Bentrem
- Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Northwestern Quality Improvement, Research, & Education in Surgery (NQUIRES), Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
7
|
Kaviyarasan V, Das A, Deka D, Saha B, Banerjee A, Sharma NR, Duttaroy AK, Pathak S. Advancements in immunotherapy for colorectal cancer treatment: a comprehensive review of strategies, challenges, and future prospective. Int J Colorectal Dis 2024; 40:1. [PMID: 39731596 DOI: 10.1007/s00384-024-04790-w] [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] [Accepted: 12/15/2024] [Indexed: 12/30/2024]
Abstract
PURPOSE Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Metastatic colorectal cancer (mCRC) continues to present significant challenges, particularly in patients with proficient mismatch repair/microsatellite stable (pMMR/MSS) tumors. This narrative review aims to provide recent developments in immunotherapy for CRC treatment, focusing on its efficacy and challenges. METHODS This review discussed the various immunotherapeutic strategies for CRC treatment, including immune checkpoint inhibitors (ICIs) targeting PD-1 and PD-L1, combination therapies involving ICIs with other modalities, chimeric antigen receptor T-cell (CAR-T) cell therapy, and cancer vaccines. The role of the tumor microenvironment and immune evasion mechanisms was also explored to understand their impact on the effectiveness of these therapies. RESULTS This review provides a comprehensive update of recent advancements in immunotherapy for CRC, highlighting the potential of various immunotherapeutic approaches, including immune checkpoint inhibitors, combination therapies, CAR-T therapy, and vaccination strategies. The results of checkpoint inhibitors, particularly in patients with MSI-H/dMMR tumors, which have significant improvements in survival rates have been observed. Furthermore, this review also addresses the challenges faced in treating pMMR/MSS CRC, which remains resistant to immunotherapy. CONCLUSION Immunotherapy plays a significant role in the treatment of CRC, particularly in patients with MSI-H/dMMR tumors. However, many challenges remain, especially in treating pMMR/MSS CRC. This review discussed the need for further research into combination therapies, biomarker development, CAR-T cell therapy, and a deeper understanding of immune evasion mechanisms for CRC treatment.
Collapse
Affiliation(s)
- Vaishak Kaviyarasan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Dikshita Deka
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Biki Saha
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| | - Neeta Raj Sharma
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
8
|
Chowdhury D, Das A, Mishra M, Khutere T, Bodakhe SH. Physiological markers for immunotherapeutics: a review. J Chemother 2024:1-24. [PMID: 39711144 DOI: 10.1080/1120009x.2024.2443701] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Immunotherapy has been advanced through multiple approaches, including immunogenic cytokines, monoclonal antibodies, therapeutic vaccinations, adoptive cell transfer, stem cell transplantation, and oncolytic viruses. This review analyses various strategies in genomics, transcriptomics, single-cell techniques, computational analysis, big data, and imaging technologies for the identification of tumour microbiota and microenvironments. Immunotherapy is becoming acknowledged as a feasible cancer treatment method, facilitating innovative cancer medicines and personalized medicine techniques.
Collapse
Affiliation(s)
- Durlav Chowdhury
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Ashmita Das
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Mrityunjay Mishra
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Trinkal Khutere
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| | - Surendra H Bodakhe
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, Chhattisgarh, India
| |
Collapse
|
9
|
Claeys C, Billiet A, Haustermans K, Wolthuis A, Bislenghi G, D'Hoore A, Dresen R, Rasschaert G, Van Cutsem E, Van Herpe F, Dekervel J. Organ Preservation After Immune Checkpoint Inhibition for Locally Advanced Rectal Cancer. Clin Colorectal Cancer 2024:S1533-0028(24)00118-X. [PMID: 39794189 DOI: 10.1016/j.clcc.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 01/13/2025]
Affiliation(s)
- Charlotte Claeys
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium
| | - Antoon Billiet
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium
| | - Karin Haustermans
- Department of Radiation Oncology, University Hospital Leuven, Leuven, Belgium
| | - Albert Wolthuis
- Department of Abdominal Surgery, University Hospital Leuven, Leuven, Belgium
| | - Gabriele Bislenghi
- Department of Abdominal Surgery, University Hospital Leuven, Leuven, Belgium
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospital Leuven, Leuven, Belgium
| | - Raphaëla Dresen
- Department of Radiology, University Hospital Leuven, Leuven, Belgium
| | - Gertjan Rasschaert
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium
| | - Eric Van Cutsem
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium
| | - Filip Van Herpe
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium
| | - Jeroen Dekervel
- Department of Gastroenterology/Digestive oncology, University Hospital Leuven, Leuven, Belgium.
| |
Collapse
|
10
|
Ahluwalia P, Mondal AK, Vashisht A, Singh H, Alptekin A, Ballur K, Omar N, Ahluwalia M, Jones K, Barrett A, Kota V, Kolhe R. Identification of a distinctive immunogenomic gene signature in stage-matched colorectal cancer. J Cancer Res Clin Oncol 2024; 151:9. [PMID: 39673574 PMCID: PMC11646222 DOI: 10.1007/s00432-024-06034-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/11/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the leading causes of cancer-related mortality worldwide. Despite advances in diagnosis and treatment, including surgery, chemotherapy, and immunotherapy, accurate clinical markers are still lacking. The development of prognostic and predictive indicators, particularly in the context of personalized medicine, could significantly improve CRC patient management. METHOD In this retrospective study, we used FFPE blocks of tissue samples from CRC patients at Augusta University (AU) to quantify a custom 15-gene panel. To differentiate the tumor and adjacent normal regions (NAT), H&E staining was utilized. For the quantification of transcripts, we used the NanoString nCounter platform. Kaplan-Meier and Log-rank tests were used to perform survival analyses. Several independent datasets were explored to validate the gene signature. Orthogonal analyses included single-cell profiling, differential gene expression, immune cell deconvolution, neoantigen prediction, and biological pathway assessment. RESULTS A 3-gene signature (GTF3A, PKM, and VEGFA) was found to be associated with overall survival in the AU cohort (HR = 2.26, 95% CI 1.05-4.84, p = 0.02, 93 patients), TCGA cohort (HR = 1.57, 95% CI 1.05-2.35, p < 0.02, 435 patients) and four other GEO datasets. Independent single-cell analysis identified relatively higher expression of the 3-gene signature in the tumor region. Differential analysis revealed dysregulated tissue inflammation, immune dysfunction, and neoantigen load of cell cycle processes among high-risk patients compared to low-risk patients. CONCLUSION We developed a 3-gene signature with the potential for prognostic and predictive clinical assessment of CRC patients. This gene-based stratification offers a cost-effective approach to personalized cancer management. Further research using similar methods could identify therapy-specific gene signatures to strengthen the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashis K Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Ahmet Alptekin
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | | | - Kimya Jones
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Amanda Barrett
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA
| | - Vamsi Kota
- Georgia Cancer Center at Augusta University, Augusta, GA, 30912, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA, 30912, BF-207, USA.
| |
Collapse
|
11
|
Selvin T, Berglund M, Åkerström A, Zia M, Rudfeldt J, Jarvius M, Larsson R, Andersson CR, Fryknäs M. Exploratory insights from the immuno-oncology hollow fiber assay: A pilot approach bridging In Vitro and In Vivo models. SLAS Technol 2024; 29:100232. [PMID: 39638255 DOI: 10.1016/j.slast.2024.100232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/13/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
To facilitate the translation of immunotherapies from bench to bedside, predictive preclinical models are essential. We developed the in vivo immuno-oncology Hollow Fiber Assay (HFA) to bridge the gap between simpler cell-based in vitro assays and more complex mouse models for immuno-oncology drug evaluation. The assay involves co-culturing human cancer cell lines or primary patient-derived cancer cells with human immune cells inside semipermeable hollow fibers. Implanted intraperitoneally in mice, the fibers captured treatment-induced immune cell-mediated cancer cell killing following treatments with aCD3 and/or IL-2, demonstrating the feasibility of the assay. Traditional models require lengthy observation periods to monitor tumor growth and treatment response. The immuno-oncology HFA enables a rapid initial in vivo evaluation of immunological agents on cancer and immune cells of human origin, addressing two of the 3Rs - reduction and refinement - in animal research.
Collapse
Affiliation(s)
- Tove Selvin
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden; Current address: Department of Immunology, Genetics and Pathology, Cancer Precision Medicine, Uppsala University, SE-75185, Uppsala, Sweden.
| | - Malin Berglund
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden
| | - Anders Åkerström
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden; Department of Clinical Chemistry and Pharmacology, Uppsala University Hospital, Uppsala, 751 85, Sweden
| | - Marco Zia
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden
| | - Jakob Rudfeldt
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden; Department of Clinical Chemistry and Pharmacology, Uppsala University Hospital, Uppsala, 751 85, Sweden
| | - Malin Jarvius
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden; Current address: Department of Pharmaceutical Biosciences and Science for Life Laboratory, Uppsala University, Box 591, SE-751 24, Uppsala, Sweden
| | - Rolf Larsson
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden
| | - Claes R Andersson
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden
| | - Mårten Fryknäs
- Department of Medical Sciences, Division of Cancer Pharmacology and Computational Medicine, Uppsala University, SE-75185, Uppsala, Sweden.
| |
Collapse
|
12
|
Pan L, Zhou Y, Kuang Y, Wang C, Wang W, Hu X, Chen X. Progress of research on γδ T cells in colorectal cancer (Review). Oncol Rep 2024; 52:160. [PMID: 39364743 PMCID: PMC11478060 DOI: 10.3892/or.2024.8819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent malignancy and second leading cause of cancer‑related fatalities worldwide. Immunotherapy alone or in combination with chemotherapy has a favorable survival benefit for patients with CRC. Unlike αβ T cells, which are prone to drug resistance, γδ T cells do not exhibit major histocompatibility complex restriction and can target tumor cells through diverse mechanisms. Recent research has demonstrated the widespread involvement of Vδ1T, Vδ2T, and γδ T17 cells in tumorigenesis and progression. In the present review, the influence of different factors, including immune checkpoint molecules, the tumor microenvironment and microorganisms, was summarized on the antitumor/protumor effects of these cells, aiming to provide insights for the development of more efficient and less toxic immunotherapy‑based anticancer drugs.
Collapse
Affiliation(s)
- Lijuan Pan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Yiru Zhou
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Yeye Kuang
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Chan Wang
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Weimin Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| | - Xiaotong Hu
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
- Department of Pathology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang 310016, P.R. China
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P.R. China
| |
Collapse
|
13
|
González-Montero J, Rojas CI, Burotto M. Predictors of response to immunotherapy in colorectal cancer. Oncologist 2024; 29:824-832. [PMID: 38920285 PMCID: PMC11449076 DOI: 10.1093/oncolo/oyae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Colorectal cancer (CRC) is a major cause of cancer-related deaths globally. While treatment advancements have improved survival rates, primarily through targeted therapies based on KRAS, NRAS, and BRAF mutations, personalized treatment strategies for CRC remain limited. Immunotherapy, mainly immune checkpoint blockade, has shown efficacy in various cancers but is effective in only a small subset of patients with CRC with deficient mismatch repair (dMMR) proteins or high microsatellite instability (MSI). Recent research has challenged the notion that CRC is immunologically inert, revealing subsets with high immunogenicity and diverse lymphocytic infiltration. Identifying precise biomarkers beyond dMMR and MSI is crucial to expanding immunotherapy benefits. Hence, exploration has extended to various biomarker sources, such as the tumor microenvironment, genomic markers, and gut microbiota. Recent studies have introduced a novel classification system, consensus molecular subtypes, that aids in identifying patients with CRC with an immunogenic profile. These findings underscore the necessity of moving beyond single biomarkers and toward a comprehensive understanding of the immunological landscape in CRC, facilitating the development of more effective, personalized therapies.
Collapse
Affiliation(s)
- Jaime González-Montero
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
- Basic and Clinical Oncology Department, University of Chile, Santiago 838045, Chile
| | - Carlos I Rojas
- Bradford Hill Clinical Research Center, Santiago 8420383, Chile
| | | |
Collapse
|
14
|
Güleç Taşkıran AE, Karaoğlu DA, Eylem CC, Ermiş Ç, Güderer İ, Nemutlu E, Demirkol Canlı S, Banerjee S. Glutamine withdrawal leads to the preferential activation of lipid metabolism in metastatic colorectal cancer. Transl Oncol 2024; 48:102078. [PMID: 39111172 PMCID: PMC11362781 DOI: 10.1016/j.tranon.2024.102078] [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: 03/31/2024] [Revised: 07/23/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
INTRODUCTION Glutamine is a non-essential amino acid that is critical for cell growth. However, the differential metabolism of l-glutamine in metastatic versus primary colorectal cancer (CRC) has not been evaluated adequately. MATERIALS AND METHODS Differential expression of glutamine-related genes was determined in primary versus metastatic CRC. Univariate Cox regression and hierarchical clustering were used to generate a gene signature for prognostication. Untargeted metabolomics and 18O based fluxomics were used to identify differential metabolite levels and energy turnover in the paired primary (SW480) and metastatic (SW620) CRC cells. Western blot and qRT-PCR were used to validate differential gene expression. Subcellular localization of E-cadherin was determined by immunocytochemistry. Lipid droplets were visualized with Nile Red. RESULTS The GO term "Glutamine metabolism" was significantly enriched in metastatic versus primary tumors. Supporting this, SW620 cells showed decreased membrane localization of E-cadherin and increased motility upon l-Glutamine withdrawal. A glutamine related signature associated with worse prognosis was identified and validated in multiple datasets. A fluxomics assay revealed a slower TCA cycle in SW480 and SW620 cells upon l-Glutamine withdrawal. SW620 cells, however, could maintain high ATP levels. Untargeted metabolomics indicated the preferential metabolism of fatty acids in SW620 but not SW480 cells. Lipids were mainly obtained from the environment rather than by de novo synthesis. CONCLUSIONS Metastatic CRC cells can display aberrant glutamine metabolism. We show for the first time that upon l-glutamine withdrawal, SW620 (but not SW480) cells were metabolically plastic and could metabolize lipids for survival and cellular motility.
Collapse
Affiliation(s)
- Aliye Ezgi Güleç Taşkıran
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye; Department of Molecular Biology and Genetics, Başkent University, Ankara, Türkiye
| | | | - Cemil Can Eylem
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Çağdaş Ermiş
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye
| | - İsmail Güderer
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Türkiye
| | - Seçil Demirkol Canlı
- Division of Tumor Pathology, Department of Clinical Oncology, Cancer Institute, Hacettepe University, Ankara, Türkiye
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi, Ankara, Türkiye; Cancer Systems Biology (CanSyL), Orta Doğu Teknik Üniversitesi, Ankara, Türkiye.
| |
Collapse
|
15
|
Al-Khazraji Y, Muzammil MA, Javid S, Tangella AV, Gohil NV, Saifullah H, Kanagala SG, Fariha F, Muneer A, Ahmed S, Shariq A. Novel regimens and treatment strategies in neoadjuvant therapy for colorectal cancer: A systematic review. Int J Health Sci (Qassim) 2024; 18:43-58. [PMID: 39282125 PMCID: PMC11393386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Objective The objective of this systematic review was to describe novel regimens and treatment strategies in neoadjuvant therapy for colorectal cancer (CRC). The aim was to summarize the current advancements in neoadjuvant chemotherapy (NACT) for CRC, including the use of cytotoxic drugs, targeted treatments, and immunotherapy. The analysis aimed to provide insights into the potential benefits and drawbacks of these novel approaches and highlight the need for further research to optimize NACT use in CRC and improve patient outcomes. Methods From October 20, 2023, to December 10, 2023, a comprehensive literature search was conducted across multiple databases, including PubMed, Ovid, Web of Science, the Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, Embase, and Scopus. Studies addressing the use of and treatment strategies for CRC and neoadjuvant therapies were included. Screening was conducted in two steps, initially by title and abstract and then by full-text articles. English-language articles were considered, while preprints, non-English publications, and articles published as grey literature were excluded from the study. A total of 85 studies were selected for further analysis after screening and filtering. Results After filtering out duplicates and items that were irrelevant to our research query from the initial database search's 510 results, 397 unique articles were found. Eighty-five studies were chosen for additional analysis after the articles underwent two rounds of screening. Conclusion The review concluded that neoadjuvant therapy for CRC has evolved beyond conventional approaches and holds promise for improving patient outcomes. Future prospects for advancing neoadjuvant approaches are promising, with ongoing clinical trials investigating the refinement of strategies, identification of predictive biomarkers, and optimization of patient selection. The adoption of novel regimens, precision medicine, and immunotherapy offers opportunities to redefine treatment paradigms and enhance patient care in CRC.
Collapse
Affiliation(s)
| | | | - Saman Javid
- Department of Medicine, CMH Kharian Medical College, Kharian, Pakistan
| | | | - Namra Vinay Gohil
- Department of Medicine, Medical College Baroda, Vadodara, Gujarat, India
| | - Hanya Saifullah
- Department of Medicine, Medical College Baroda, CMH Lahore Medical College, Lahore, Pakistan
| | | | - Fnu Fariha
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Asim Muneer
- Department of Adult Hematology Oncology, Prince Faisal Ca ncer Centre Buraidah, Al qaseem, Saudi Arabia
| | - Sumaira Ahmed
- Department of Gastroenterology, King Fahad Hospital, Burydah, KSA
| | - Ali Shariq
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
16
|
Chen C, Luo X, Tang W, Geng H, Martínez-Pérez J, Price T, Kang L, Lu H, Zhang Y. Regorafenib combined with immune checkpoint inhibitors versus regorafenib monotherapy as a late-line treatment for metastatic colorectal cancer: a single-center, retrospective cohort study. J Gastrointest Oncol 2024; 15:1497-1507. [PMID: 39279938 PMCID: PMC11399868 DOI: 10.21037/jgo-24-468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Background Few data are available on metastatic colorectal cancer (mCRC) treated with late-line regorafenib monotherapy or combined with other therapies. This study thus aimed to examine regorafenib combined with immune checkpoint inhibitors (ICIs) compared with regorafenib monotherapy in patients with advanced CRC. Methods This single-center retrospective cohort study included patients with advanced CRC who experienced recurrence and progression after standard first- and second-line treatments treatment from November 2018 to December 2021. The patients received regorafenib plus ICIs or regorafenib monotherapy. Treatment response was evaluated based on Response Evaluation Criteria in Solid Tumors (RECIST). Overall survival (OS) and progression-free survival (PFS) were analyzed via multivariate analysis. Results The combined group and the monotherapy group included 30 and 43 patients, respectively. The median OS (13.7 vs. 10.1 months; P=0.10) and PFS (4 vs. 3.6 months; P=0.32) were not significantly different between the two groups. In males, the median OS was significantly longer in the combined group compared with the monotherapy group (not reached vs. 8.03 months; P=0.02), but the median PFS showed no significant difference (7.23 vs. 3.90 months; P=0.16). There was no significant difference in OS (P=0.71) or PFS (P=0.89) in females. Eastern Cooperative Oncology Group performance status (ECOG PS) 1 [vs. 0; hazard ratio (HR) =3.13, 95% confidence interval (CI): 1.61-6.10; P<0.001] was independently associated with PFS. ECOG PS 1 (vs. 0; HR =3.63, 95% CI: 1.54-8.56; P=0.003) and combined therapy (vs. monotherapy; HR =0.47, 95% CI: 0.22-0.99; P=0.048) were associated with OS. Conclusions Regorafenib combined with ICIs led to numerically longer PFS and significantly prolonged OS in patients with mCRC compared to regorafenib monotherapy, especially in male patients.
Collapse
Affiliation(s)
- Can Chen
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Xi Luo
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Wenhua Tang
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Haofei Geng
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Julia Martínez-Pérez
- Medical Oncology Department, University Hospital Virgen del Rocio, Seville, Spain
- Institute of Biomedicine of Seville (IBIS) (HUVR/University of Sevilla/CSIC), Seville, Spain
| | - Timothy Price
- Department of Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
- University of Adelaide, School of Medicine, Adelaide, Australia
| | - Lili Kang
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Honglian Lu
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| | - Yanling Zhang
- Department of Oncology, The First Affiliated Hospital of the Army Medical University, Chongqing, China
| |
Collapse
|
17
|
Manriquez E, Solé S, Silva J, Hermosilla JP, Romero R, Quezada-Diaz F. Deciphering the Dilemma: Choosing the Optimal Total Neoadjuvant Treatment Strategy for Locally Advanced Rectal Cancer. Curr Oncol 2024; 31:4292-4304. [PMID: 39195303 PMCID: PMC11352987 DOI: 10.3390/curroncol31080320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/12/2024] [Accepted: 07/27/2024] [Indexed: 08/29/2024] Open
Abstract
Rectal cancer management has evolved significantly, particularly with neoadjuvant treatment strategies. This narrative review examines the development and effectiveness of these therapies for locally advanced rectal cancer (LARC), highlighting the historical quest that led to current neoadjuvant alternatives. Initially, trials showed the benefits of adding radiotherapy (RT) and chemotherapy (CT) to surgery, reducing local recurrence (LR). The addition of oxaliplatin to chemoradiotherapy (CRT) further improved outcomes. TNT integrates chemotherapy and radiotherapy preoperatively to enhance adherence, timing, and systemic control. Key trials, including PRODIGE 23, CAO/ARO/AIO 12, OPRA, RAPIDO, and STELLAR, are analyzed to compare short-course and long-course RT with systemic chemotherapy. The heterogeneity and difficulty in comparing TNT trials due to different designs and outcomes are acknowledged, along with their promising long-term results. On the other hand, it briefly discusses the potential for non-operative management (NOM) in select patients, a strategy gaining traction due to favorable outcomes in specific trials. As a conclusion, this review underscores the complexity of rectal cancer treatment, emphasizing individualized approaches considering patient preferences and healthcare resources. It also highlights the importance of interpreting impressive positive or negative results with caution due to the variability in study designs and patient populations.
Collapse
Affiliation(s)
- Erik Manriquez
- Complejo Asistencial Doctor Sotero del Rio, Santiago 8207257, Chile;
| | - Sebastián Solé
- Clínica IRAM, Santiago 7630370, Chile; (S.S.); (J.S.)
- Radiotherapy Oncology Program, School of Medicine, Universidad Diego Portales, Santiago 8370191, Chile; (J.P.H.); (R.R.)
| | - Javiera Silva
- Clínica IRAM, Santiago 7630370, Chile; (S.S.); (J.S.)
- Radiotherapy Oncology Program, School of Medicine, Universidad Diego Portales, Santiago 8370191, Chile; (J.P.H.); (R.R.)
| | - Juan Pablo Hermosilla
- Radiotherapy Oncology Program, School of Medicine, Universidad Diego Portales, Santiago 8370191, Chile; (J.P.H.); (R.R.)
- Hospital Dipreca, Santiago 7601003, Chile
| | - Rubén Romero
- Radiotherapy Oncology Program, School of Medicine, Universidad Diego Portales, Santiago 8370191, Chile; (J.P.H.); (R.R.)
- Hospital Dipreca, Santiago 7601003, Chile
| | - Felipe Quezada-Diaz
- Complejo Asistencial Doctor Sotero del Rio, Santiago 8207257, Chile;
- Clínica Universidad de los Andes, Santiago 7620157, Chile
- Center for Cancer Prevention and Control (CECAN), Santiago 8380453, Chile
| |
Collapse
|
18
|
Cheng CC, Chang J, Ho AS, Sie ZL, Peng CL, Wang CL, Dev K, Chang CC. Tumor-intrinsic IFNα and CXCL10 are critical for immunotherapeutic efficacy by recruiting and activating T lymphocytes in tumor microenvironment. Cancer Immunol Immunother 2024; 73:175. [PMID: 38953994 PMCID: PMC11219622 DOI: 10.1007/s00262-024-03761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 06/13/2024] [Indexed: 07/04/2024]
Abstract
Tumor immunotherapies targeting PD-(L)1 exhibit anti-tumor efficacy in only 10-30% of patients with various cancers. Literature has demonstrated that a "hot tumor" which contains high T lymphocytes in the tumor microenvironment exhibits a better response to immunotherapies than a "cold tumor." This study aimed to investigate whether tumor-intrinsic IFNα and CXCL10 determine the recruitment and activation of CD8+ T cells to become "hot tumor." In this study, we found that CXCL10 overexpressed in a variety of tumors including lung, colon, and liver tumors with a correlation with PD-L1. High PD-L1 and CXCL10 are associated with better survival rates in tumor patients receiving immunotherapies. IFNs-downstream transcriptional factor IRF-1 and STAT1 were correlated with PD-L1 and CXCL10 expression. We demonstrated that IRF-1 and STAT1 were both bound with the promoters of PD-L1 and CXCL10, sharing the same signaling pathway and determining IFNs-mediated PD-L1 and CXCL10 expression. In addition, IFNα significantly increased activation marker IFNγ in PBMCs, promoting M1 type monocyte differentiation, CD4+ T, and CD8+ T cell activation. Particularly, we found that CD8+ T lymphocytes abundantly expressed CXCR3, a receptor of CXCL10, by flow cytometry, indicating that tumor-intrinsic CXCL10 potentially recruited CD8+ T in tumor microenvironment. To demonstrate the hypothesis, immunotherapy-sensitive CT26 and immunotherapy-resistant LL/2 were used and we found that CT26 cells exhibited higher IFNα, IFNγ, CXCL10, and PD-L1 levels compared to LL/2, leading to higher IFNγ expression in mouse splenocytes. Moreover, we found that CD8+ T cells were recruited by CXCL10 in vitro, whereas SCH546738, an inhibitor of CXCR3, inhibited T cell migration and splenocytes-mediated anti-tumor effect. We then confirmed that CT26-derived tumor was sensitive to αPD-L1 immunotherapy and LL/2-tumor was resistant, whereas αPD-L1 significantly increased T lymphocyte activation marker CD107a in CT26-derived BALB/c mice. In conclusion, this study revealed that CXCL10 expression is correlated with PD-L1 in tumors, sharing the same signaling pathway and associating with better immunotherapeutic efficacy. Further evidence in the syngeneic tumor models demonstrated that immunotherapy-sensitive CT26 intrinsically exhibited higher IFNα and CXCL10 compared to immunotherapy-resistant LL/2 to recruit and activate CD8+ T cells in the tumor microenvironment, exhibiting "hot tumor" characteristic of sensitizing αPD-L1 immunotherapies.
Collapse
Affiliation(s)
- Chun-Chia Cheng
- Research Center of Radiation Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, 333, Taiwan
| | - Jungshan Chang
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan
| | - Ai-Sheng Ho
- Division of Gastroenterology, Cheng Hsin General Hospital, Taipei, 112, Taiwan
| | - Zong-Lin Sie
- Research Center of Radiation Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Cheng-Liang Peng
- Department of Isotope Application Research, National Atomic Research Institute, Taoyuan, 325, Taiwan
| | - Chih-Liang Wang
- Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Linkou, Taoyuan, 333, Taiwan
| | - Kapil Dev
- Research Center of Radiation Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei, 110, Taiwan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 110, Taiwan.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, 110, Taiwan.
| |
Collapse
|
19
|
Teng HW, Wang TY, Lin CC, Tong ZJ, Cheng HW, Wang HT. Interferon Gamma Induces Higher Neutrophil Extracellular Traps Leading to Tumor-Killing Activity in Microsatellite Stable Colorectal Cancer. Mol Cancer Ther 2024; 23:1043-1056. [PMID: 38346939 DOI: 10.1158/1535-7163.mct-23-0744] [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: 11/14/2023] [Revised: 01/18/2024] [Accepted: 02/05/2024] [Indexed: 07/03/2024]
Abstract
Many patients with colorectal cancer do not respond to immune checkpoint blockade (ICB) therapy, highlighting the urgent need to understand tumor resistance mechanisms. Recently, the link between the IFNγ signaling pathway integrity and ICB resistance in the colorectal cancer tumor microenvironment has been revealed. The immunosuppressive microenvironment poses a significant challenge to antitumor immunity in colorectal cancer development. Tumor-associated neutrophils found in tumor tissues exhibit an immunosuppressive phenotype and are associated with colorectal cancer patient prognosis. Neutrophil extracellular traps (NET), DNA meshes containing cytotoxic enzymes released into the extracellular space, may be promising therapeutic targets in cancer. This study showed increased NETs in tumor tissues and peripheral neutrophils of high levels of microsatellite instability (MSI-H) patients with colorectal cancer compared with microsatellite stable (MSS) patients with colorectal cancer. IFNγ response genes were enriched in MSI-H patients with colorectal cancer compared with patients with MSS colorectal cancer. Co-culturing neutrophils with MSI-H colorectal cancer cell lines induced more NET formation and higher cellular apoptosis than MSS colorectal cancer cell lines. IFNγ treatment induced more NET formation and apoptosis in MSS colorectal cancer cell lines. Using subcutaneous or orthotopic CT-26 (MSS) tumor-bearing mice models, IFNγ reduced tumor size and enhanced PD-1 antibody-induced tumor-killing activity, accompanied by upregulated NETs and cellular apoptosis. These findings suggest that IFNγ could be a therapeutic strategy for MSS colorectal cancer.
Collapse
Affiliation(s)
- Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tean-Ya Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Chi Lin
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Colon and Rectal Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zhen-Jie Tong
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiao-Wei Cheng
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsiang-Tsui Wang
- Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Doctor degree program in Toxicology, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
O'Connell RM, Hoti E. Challenges and Opportunities for Precision Surgery for Colorectal Liver Metastases. Cancers (Basel) 2024; 16:2379. [PMID: 39001441 PMCID: PMC11240734 DOI: 10.3390/cancers16132379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The incidence of colorectal cancer and colorectal liver metastases (CRLM) is increasing globally due to an interaction of environmental and genetic factors. A minority of patients with CRLM have surgically resectable disease, but for those who have resection as part of multimodal therapy for their disease, long-term survival has been shown. Precision surgery-the idea of careful patient selection and targeting of surgical intervention, such that treatments shown to be proven to benefit on a population level are the optimal treatment for each individual patient-is the new paradigm of care. Key to this is the understanding of tumour molecular biology and clinically relevant mutations, such as KRAS, BRAF, and microsatellite instability (MSI), which can predict poorer overall outcomes and a poorer response to systemic therapy. The emergence of immunotherapy and hepatic artery infusion (HAI) pumps show potential to convert previously unresectable disease to resectable disease, in addition to established systemic and locoregional therapies, but the surgeon must be wary of poor-quality livers and the spectre of post-hepatectomy liver failure (PHLF). Volume modulation, a cornerstone of hepatic surgery for a generation, has been given a shot in the arm with the advent of liver venous depletion (LVD) ensuring significantly more hypertrophy of the future liver remnant (FLR). The optimal timing of liver resection for those patients with synchronous disease is yet to be truly established, but evidence would suggest that those patients requiring complex colorectal surgery and major liver resection are best served with a staged approach. In the operating room, parenchyma-preserving minimally invasive surgery (MIS) can dramatically reduce the surgical insult to the patient and lead to better perioperative outcomes, with quicker return to function.
Collapse
Affiliation(s)
- Robert Michael O'Connell
- Department of Hepatopancreaticobiliary and Transplantation Surgery, Saint Vincent's University Hospital, D04 T6F4 Dublin, Ireland
| | - Emir Hoti
- Department of Hepatopancreaticobiliary and Transplantation Surgery, Saint Vincent's University Hospital, D04 T6F4 Dublin, Ireland
| |
Collapse
|
21
|
Abedizadeh R, Majidi F, Khorasani HR, Abedi H, Sabour D. Colorectal cancer: a comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev 2024; 43:729-753. [PMID: 38112903 DOI: 10.1007/s10555-023-10158-3] [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: 08/08/2023] [Accepted: 11/27/2023] [Indexed: 12/21/2023]
Abstract
Colorectal cancer is the third most common and the second deadliest cancer worldwide. To date, colorectal cancer becomes one of the most important challenges of the health system in many countries. Since the clinical symptoms of this cancer appear in the final stages of the disease and there is a significant golden time between the formation of polyps and the onset of cancer, early diagnosis can play a significant role in reducing mortality. Today, in addition to colonoscopy, minimally invasive methods such as liquid biopsy have received much attention. The treatment of this complex disease has been mostly based on traditional treatments including surgery, radiotherapy, and chemotherapy; the high mortality rate indicates a lack of success for current treatment methods. Moreover, disease recurrence is another problem of traditional treatments. Recently, new approaches such as targeted therapy, immunotherapy, and nanomedicine have opened new doors for cancer treatment, some of which have already entered the market, and many methods have shown promising results in clinical trials. The success of immunotherapy in the treatment of refractory disease, the introduction of these methods into neoadjuvant therapy, and the successful results in tumor shrinkage without surgery have made immunotherapy a tough competitor for conventional treatments. It seems that the combination of those methods with such targeted therapies will go through promising changes in the future of colorectal cancer treatment.
Collapse
Affiliation(s)
- Roya Abedizadeh
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Fateme Majidi
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hamid Reza Khorasani
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran
| | - Hassan Abedi
- Department of Internal Medicine, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran.
| | - Davood Sabour
- Department of Cancer Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Isar 11, Babol, 47138-18983, Iran.
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Bani-Hashem Square, Tehran, 16635-148, Iran.
| |
Collapse
|
22
|
Nikolouzakis TK, Chrysos E, Docea AO, Fragkiadaki P, Souglakos J, Tsiaoussis J, Tsatsakis A. Current and Future Trends of Colorectal Cancer Treatment: Exploring Advances in Immunotherapy. Cancers (Basel) 2024; 16:1995. [PMID: 38893120 PMCID: PMC11171065 DOI: 10.3390/cancers16111995] [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: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer of the colon and rectum (CRC) has been identified among the three most prevalent types of cancer and cancer-related deaths for both sexes. Even though significant progress in surgical and chemotherapeutic techniques has markedly improved disease-free and overall survival rates in contrast to those three decades ago, recent years have seen a stagnation in these improvements. This underscores the need for new therapies aiming to augment patient outcomes. A number of emerging strategies, such as immune checkpoint inhibitors (ICIs) and adoptive cell therapy (ACT), have exhibited promising outcomes not only in preclinical but also in clinical settings. Additionally, a thorough appreciation of the underlying biology has expanded the scope of research into potential therapeutic interventions. For instance, the pivotal role of altered telomere length in early CRC carcinogenesis, leading to chromosomal instability and telomere dysfunction, presents a promising avenue for future treatments. Thus, this review explores the advancements in CRC immunotherapy and telomere-targeted therapies, examining potential synergies and how these novel treatment modalities intersect to potentially enhance each other's efficacy, paving the way for promising future therapeutic advancements.
Collapse
Affiliation(s)
| | - Emmanuel Chrysos
- Department of General Surgery, University General Hospital of Heraklion, 71110 Heraklion, Greece; (T.K.N.); (E.C.)
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Persefoni Fragkiadaki
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| | - John Souglakos
- Laboratory of Translational Oncology, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - John Tsiaoussis
- Department of Anatomy, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece; (P.F.); (A.T.)
| |
Collapse
|
23
|
Pincus MR, Silberstein M, Zohar N, Sarafraz-Yazdi E, Bowne WB. Poptosis or Peptide-Induced Transmembrane Pore Formation: A Novel Way to Kill Cancer Cells without Affecting Normal Cells. Biomedicines 2024; 12:1144. [PMID: 38927351 PMCID: PMC11201261 DOI: 10.3390/biomedicines12061144] [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: 03/04/2024] [Revised: 04/15/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advances in cancer treatment like personalized chemotherapy and immunotherapy are aimed at tumors that meet certain specifications. In this review, we describe a new approach to general cancer treatment, termed peptide-induced poptosis, in which specific peptides, e.g., PNC-27 and its shorter analogue, PNC-28, that contain the segment of the p53 transactivating 12-26 domain that bind to HDM-2 in its 1-109 domain, bind to HDM-2 in the membranes of cancer cells, resulting in transmembrane pore formation and the rapid extrusion of cancer cell contents, i.e., tumor cell necrosis. These peptides cause tumor cell necrosis of a wide variety of solid tissue and hematopoietic tumors but have no effect on the viability and growth of normal cells since they express at most low levels of membrane-bound HDM-2. They have been found to successfully treat a highly metastatic pancreatic tumor as well as stem-cell-enriched human acute myelogenous leukemias in nude mice, with no evidence of off-target effects. These peptides also are cytotoxic to chemotherapy-resistant cancers and to primary tumors. We performed high-resolution scanning immuno-electron microscopy and visualized the pores in cancer cells induced by PNC-27. This peptide forms 1:1 complexes with HDM-2 in a temperature-independent step, followed by dimerization of these complexes to form transmembrane channels in a highly temperature-dependent step parallel to the mode of action of other membranolytic but less specific agents like streptolysin. These peptides therefore may be effective as general anti-cancer agents.
Collapse
Affiliation(s)
- Matthew R. Pincus
- Department of Pathology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | | | - Nitzan Zohar
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.Z.); (W.B.B.)
| | | | - Wilbur B. Bowne
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer, Thomas Jefferson University, Philadelphia, PA 19107, USA; (N.Z.); (W.B.B.)
| |
Collapse
|
24
|
Gharib E, Rejali L, Piroozkhah M, Zonoobi E, Nasrabadi PN, Arabsorkhi Z, Baghdar K, Shams E, Sadeghi A, Kuppen PJK, Salehi Z, Nazemalhosseini-Mojarad E. IL-2RG as a possible immunotherapeutic target in CRC predicting poor prognosis and regulated by miR-7-5p and miR-26b-5p. J Transl Med 2024; 22:439. [PMID: 38720389 PMCID: PMC11080123 DOI: 10.1186/s12967-024-05251-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Despite advances in treatment strategies, colorectal cancer (CRC) continues to cause significant morbidity and mortality, with mounting evidence a close link between immune system dysfunctions issued. Interleukin-2 receptor gamma (IL-2RG) plays a pivotal role as a common subunit receptor in the IL-2 family cytokines and activates the JAK-STAT pathway. This study delves into the role of Interleukin-2 receptor gamma (IL-2RG) within the tumor microenvironment and investigates potential microRNAs (miRNAs) that directly inhibit IL-2RG, aiming to discern their impact on CRC clinical outcomes. Bioinformatics analysis revealed a significant upregulation of IL-2RG mRNA in TCGA-COAD samples and showed strong correlations with the infiltration of various lymphocytes. Single-cell analysis corroborated these findings, highlighting IL-2RG expression in critical immune cell subsets. To explore miRNA involvement in IL-2RG dysregulation, mRNA was isolated from the tumor tissues and lymphocytes of 258 CRC patients and 30 healthy controls, and IL-2RG was cloned into the pcDNA3.1/CT-GFP-TOPO vector. Human embryonic kidney cell lines (HEK-293T) were transfected with this construct. Our research involved a comprehensive analysis of miRPathDB, miRWalk, and Targetscan databases to identify the miRNAs associated with the 3' UTR of human IL-2RG. The human microRNA (miRNA) molecules, hsa-miR-7-5p and hsa-miR-26b-5p, have been identified as potent suppressors of IL-2RG expression in CRC patients. Specifically, the downregulation of hsa-miR-7-5p and hsa-miR-26b-5p has been shown to result in the upregulation of IL-2RG mRNA expression in these patients. Prognostic evaluation of IL-2RG, hsa-miR-7-5p, and hsa-miR-26b-5p, using TCGA-COAD data and patient samples, established that higher IL-2RG expression and lower expression of both miRNAs were associated with poorer outcomes. Additionally, this study identified several long non-coding RNAs (LncRNAs), such as ZFAS1, SOX21-AS1, SNHG11, SNHG16, SNHG1, DLX6-AS1, GAS5, SNHG6, and MALAT1, which may act as competing endogenous RNA molecules for IL2RG by sequestering shared hsa-miR-7-5p and hsa-miR-26b-5p. In summary, this investigation underscores the potential utility of IL-2RG, hsa-miR-7-5p, and hsa-miR-26b-5p as serum and tissue biomarkers for predicting CRC patient prognosis while also offering promise as targets for immunotherapy in CRC management.
Collapse
Affiliation(s)
- Ehsan Gharib
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Leili Rejali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elham Zonoobi
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Parinaz Nasri Nasrabadi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Arabsorkhi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghdar
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elahe Shams
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman Street, Chamran Expressway, P.O. Box: 19857-17411, Tehran, Iran
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ehsan Nazemalhosseini-Mojarad
- Department of Surgery, Leiden University Medical Center, Leiden, Netherlands.
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman Street, Chamran Expressway, P.O. Box: 19857-17411, Tehran, Iran.
| |
Collapse
|
25
|
Ahmad Zawawi SS, Salleh EA, Musa M. Spheroids and organoids derived from colorectal cancer as tools for in vitro drug screening. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:409-431. [PMID: 38745769 PMCID: PMC11090692 DOI: 10.37349/etat.2024.00226] [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: 12/13/2023] [Accepted: 02/02/2024] [Indexed: 05/16/2024] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease. Conventional two-dimensional (2D) culture employing cell lines was developed to study the molecular properties of CRC in vitro. Although these cell lines which are isolated from the tumor niche in which cancer develop, the translation to human model such as studying drug response is often hindered by the inability of cell lines to recapture original tumor features and the lack of heterogeneous clinical tumors represented by this 2D model, differed from in vivo condition. These limitations which may be overcome by utilizing three-dimensional (3D) culture consisting of spheroids and organoids. Over the past decade, great advancements have been made in optimizing culture method to establish spheroids and organoids of solid tumors including of CRC for multiple purposes including drug screening and establishing personalized medicine. These structures have been proven to be versatile and robust models to study CRC progression and deciphering its heterogeneity. This review will describe on advances in 3D culture technology and the application as well as the challenges of CRC-derived spheroids and organoids as a mode to screen for anticancer drugs.
Collapse
Affiliation(s)
| | - Elyn Amiela Salleh
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Marahaini Musa
- Human Genome Centre, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| |
Collapse
|
26
|
Sharma S, Singh N, Turk AA, Wan I, Guttikonda A, Dong JL, Zhang X, Opyrchal M. Molecular insights into clinical trials for immune checkpoint inhibitors in colorectal cancer: Unravelling challenges and future directions. World J Gastroenterol 2024; 30:1815-1835. [PMID: 38659481 PMCID: PMC11036501 DOI: 10.3748/wjg.v30.i13.1815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 04/03/2024] Open
Abstract
Colorectal cancer (CRC) is a complex disease with diverse etiologies and clinical outcomes. Despite considerable progress in development of CRC therapeutics, challenges remain regarding the diagnosis and management of advanced stage metastatic CRC (mCRC). In particular, the five-year survival rate is very low since mCRC is currently rarely curable. Over the past decade, cancer treatment has significantly improved with the introduction of cancer immunotherapies, specifically immune checkpoint inhibitors. Therapies aimed at blocking immune checkpoints such as PD-1, PD-L1, and CTLA-4 target inhibitory pathways of the immune system, and thereby enhance anti-tumor immunity. These therapies thus have shown promising results in many clinical trials alone or in combination. The efficacy and safety of immunotherapy, either alone or in combination with CRC, have been investigated in several clinical trials. Clinical trials, including KEYNOTE-164 and CheckMate 142, have led to Food and Drug Administration approval of the PD-1 inhibitors pembrolizumab and nivolumab, respectively, for the treatment of patients with unresectable or metastatic microsatellite instability-high or deficient mismatch repair CRC. Unfortunately, these drugs benefit only a small percentage of patients, with the benefits of immunotherapy remaining elusive for the vast majority of CRC patients. To this end, primary and secondary resistance to immunotherapy remains a significant issue, and further research is necessary to optimize the use of immunotherapy in CRC and identify biomarkers to predict the response. This review provides a comprehensive overview of the clinical trials involving immune checkpoint inhibitors in CRC. The underlying rationale, challenges faced, and potential future steps to improve the prognosis and enhance the likelihood of successful trials in this field are discussed.
Collapse
Affiliation(s)
- Samantha Sharma
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Naresh Singh
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Anita Ahmed Turk
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Isabella Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Akshay Guttikonda
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Julia Lily Dong
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Xinna Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Mateusz Opyrchal
- Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
- Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|
27
|
Privitera GF, Alaimo S, Caruso A, Ferro A, Forte S, Pulvirenti A. TMBcalc: a computational pipeline for identifying pan-cancer Tumor Mutational Burden gene signatures. Front Genet 2024; 15:1285305. [PMID: 38645485 PMCID: PMC11026579 DOI: 10.3389/fgene.2024.1285305] [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: 09/04/2023] [Accepted: 03/11/2024] [Indexed: 04/23/2024] Open
Abstract
Background In the precision medicine era, identifying predictive factors to select patients most likely to benefit from treatment with immunological agents is a crucial and open challenge in oncology. Methods This paper presents a pan-cancer analysis of Tumor Mutational Burden (TMB). We developed a novel computational pipeline, TMBcalc, to calculate the TMB. Our methodology can identify small and reliable gene signatures to estimate TMB from custom targeted-sequencing panels. For this purpose, our pipeline has been trained on top of 17 cancer types data obtained from TCGA. Results Our results show that TMB, computed through the identified signature, strongly correlates with TMB obtained from whole-exome sequencing (WES). Conclusion We have rigorously analyzed the effectiveness of our methodology on top of several independent datasets. In particular we conducted a comprehensive testing on: (i) 126 samples sourced from the TCGA database; few independent whole-exome sequencing (WES) datasets linked to colon, breast, and liver cancers, all acquired from the EGA and the ICGC Data Portal. This rigorous evaluation clearly highlights the robustness and practicality of our approach, positioning it as a promising avenue for driving substantial progress within the realm of clinical practice.
Collapse
Affiliation(s)
- Grete Francesca Privitera
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Salvatore Alaimo
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Anna Caruso
- Department of Physics and Astronomy, University of Catania, Catania, Italy
| | - Alfredo Ferro
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| | - Stefano Forte
- Istituto Oncologico del Mediterraneo (IOM) Ricerca, Viagrande, Italy
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, Bioinformatics Unit, University of Catania, Catania, Italy
| |
Collapse
|
28
|
Paliogiannis P, Lobrano R, Bella MA, Fara A, Uras MG, Pinna MA, Tedde A, Madonia M, Zinellu A, Cossu A. PD-L1 immunohistochemical expression in bladder urothelial cancer with SP263, SP142 and 22C3 antibodies: A comparative study. Ann Diagn Pathol 2024; 69:152267. [PMID: 38266544 DOI: 10.1016/j.anndiagpath.2024.152267] [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: 01/13/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
Programmed death ligand 1 (PD-L1) is currently the only biomarker used for the selection of patients with bladder urothelial cancer for immunotherapy. Several platforms, antibodies and scores are currently available for the evaluation of the expression of PD-L1 in immunohistochemistry (IHC). In this study three different antibodies (SP263, SP142 and 22C3) were compared to establish their performances and concordance rates. Twenty-four consecutive cases of surgically resected urothelial cancers of the bladder were enrolled. All cases were revised, and appropriate tumor areas were selected for IHC. Three commercially available PD-L1 antibodies were tested: 22C3 pharmDx with Dako Autostainer Link 48 (Dako, Carpinteria, Ca), and SP263 and SP142 with the Ventana BenchMark (Ventana Medical Systems, Tucson, AZ) platform. All slides were evaluated by an expert pathologist and both the tumor proportion score (TPS) and the combined positive score (CPS) were determined and compared at two different cut-off levels (≥ 1 and ≥ 10). The SP263 and 22C3 clones produced more positive results with the CPS and TPS scores, respectively. The CPS score identified more positive cases than the TPS score, irrespectively of the clone or the cut-off used; the difference was statistically significant in both the SP263 and SP142 clones with the ≥1 cut-off. No statistically significant differences were found between the clones when the ≥1 cut-off was used, irrespectively of the score. At the contrary, a statistically significant difference (p = 0.024) and a trend to significance (p = 0.082) were respectively found for the TPS and CPS scores, when the SP22C3 and the SP142 clones were compared at a cut-off level of ≥10. The ICC test using CPS was 0.676 and 0.578 for the ≥1 and ≥ 10 cut-offs respectively, and 0.729 and 0.467 respectively for the same cut-offs using TPS. This suggests that the three antibodies under investigation cannot be used interchangeably, especially the 22C3 and SP142 clones which showed statistically significant difference when TPS was tested at a ≥ 10 cut-off.
Collapse
Affiliation(s)
- Panagiotis Paliogiannis
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy; Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy.
| | - Renato Lobrano
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Michele Angelo Bella
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Antonella Fara
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Maria Gabriela Uras
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy; Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| | - Maria Antonia Pinna
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Alessandro Tedde
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy; Unit of Urology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Massimo Madonia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy; Unit of Urology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy
| | - Angelo Zinellu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Antonio Cossu
- Unit of Anatomic Pathology and Histology, University Hospital of Sassari (A.O.U. SS), Sassari, Italy; Department of Medicine, Surgery and Pharmacy, University of Sassari, Sassari, Italy
| |
Collapse
|
29
|
Ulanja MB, Asafo‐Agyei KO, Neelam V, Beutler BD, Antwi‐Amoabeng D, Governor SB, Rahman GA, Djankpa FT, Ulanja RN, Nteim GB, Mabrouk T, Amankwah M, Alese OB. Survival trends for left and right sided colon cancer using population-based SEER database: A forty-five-year analysis from 1975 to 2019. Cancer Med 2024; 13:e7145. [PMID: 38651190 PMCID: PMC11036079 DOI: 10.1002/cam4.7145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/13/2024] [Accepted: 03/17/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Survival differences between left-sided colon cancer (LSCC) and right-sided colon cancer (RSCC) has been previously reported with mixed results, with various study periods not accounting for other causes of mortality. PURPOSE We sought to assess the trends in colon cancer cause- specific survival (CSS) and overall survival (OS) based on sidedness. METHOD Fine-Gray competing risk and Cox models were used to analyze Surveillance, Epidemiology, and End Results (SEER) population-based cohort from 1975 to 2019. Various interval periods were identified based on the timeline of clinical adoption of modern chemotherapy (1975-1989, interval period A; 1990-2004, B; and 2005-2019, C). RESULTS Of the 227,637 patients, 50.1% were female and 46.2% were RSCC. RSCC was more common for African Americans (51.5%), older patients (age ≥65; 51.4%), females (50.4%), while LSCC was more common among Whites (53.1%; p < 0.001), younger patients (age 18-49, 64.6%; 50-64, 62.3%; p < 0.001), males (58.1%; p < 0.001). The Median CSS for LSCC and RCC were 19.3 and 16.7 years respectively for interval period A (1975-1989). Median CSS for interval periods B and C were not reached (more than half of the cohort was still living at the end of the follow-up period). Adjusted CSS was superior for LSCC versus RSCC for the most recent interval period C (HR 0.89; 0.86-0.92; p < 0.001). LSCC consistently showed superior OS for all study periods. Stage stratification showed worse CSS for localized and regional LSCC in the earlier study periods, but the risk attenuated over time. However, left sided distant disease had superior CSS per stage for all interval periods. OS was better for LSCC irrespective of stage, with gradual improvement over time. CONCLUSION LSCC was associated with superior survival compared to right sided tumors. With the adoption of modern chemotherapy regimens, prognosis between LSCC and RSCC became more divergent in favor of LSCC. Colon cancer clinical trials should strongly consider tumor sidedness as an enrollment factor.
Collapse
Affiliation(s)
- Mark B. Ulanja
- CHRISTUS Ochsner St. Patrick HospitalLake CharlesLouisianaUSA
| | | | - Vijay Neelam
- CHRISTUS Ochsner St. Patrick HospitalLake CharlesLouisianaUSA
| | - Bryce D. Beutler
- Department of Radiology, Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | | | - Samuel B. Governor
- Saint Louis University College for Public Health and Social JusticeSaint LouisMissouriUSA
| | - Ganiyu A. Rahman
- Department of Surgery, School of Medical SciencesUniversity of Cape CoastCape CoastGhana
| | - Francis T. Djankpa
- Department of Physiology, School of Medical SciencesUniversity of Cape CoastCape CoastGhana
| | - Reginald N. Ulanja
- Department of Physiology, School of Medical SciencesUniversity of Cape CoastCape CoastGhana
| | - Grace B. Nteim
- Department of Physiology, School of Medical SciencesUniversity of Cape CoastCape CoastGhana
| | - Tarig Mabrouk
- CHRISTUS Ochsner St. Patrick HospitalLake CharlesLouisianaUSA
| | - Millicent Amankwah
- Department of Hematology Oncology, Feist‐Weiller Cancer CenterLouisiana State University Health ShreveportLouisianaUSA
| | - Olatunji B. Alese
- Department of Hematology and OncologyWinship Cancer Institute, Emory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
30
|
Shebbo S, Binothman N, Darwaish M, Niaz HA, Abdulal RH, Borjac J, Hashem AM, Mahmoud AB. Redefining the battle against colorectal cancer: a comprehensive review of emerging immunotherapies and their clinical efficacy. Front Immunol 2024; 15:1350208. [PMID: 38533510 PMCID: PMC10963412 DOI: 10.3389/fimmu.2024.1350208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer globally and presents a significant challenge owing to its high mortality rate and the limitations of traditional treatment options such as surgery, radiotherapy, and chemotherapy. While these treatments are foundational, they are often poorly effective owing to tumor resistance. Immunotherapy is a groundbreaking alternative that has recently emerged and offers new hope for success by exploiting the body's own immune system. This article aims to provide an extensive review of clinical trials evaluating the efficacy of various immunotherapies, including CRC vaccines, chimeric antigen receptor T-cell therapies, and immune checkpoint inhibitors. We also discuss combining CRC vaccines with monoclonal antibodies, delve into preclinical studies of novel cancer vaccines, and assess the impact of these treatment methods on patient outcomes. This review seeks to provide a deeper understanding of the current state of CRC treatment by evaluating innovative treatments and their potential to redefine the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Salima Shebbo
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Najat Binothman
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Chemistry, College of Sciences and Arts, King Abdulaziz University, Rabigh, Saudi Arabia
| | - Manar Darwaish
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Hanan A. Niaz
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Rwaa H. Abdulal
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jamilah Borjac
- Department of Biological Sciences, Beirut Arab University, Debbieh, Lebanon
| | - Anwar M. Hashem
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmad Bakur Mahmoud
- Strategic Research and Innovation Laboratories, Taibah University, Madinah, Saudi Arabia
- College of Applied Medical Sciences, Taibah University, Almadinah Almunawarah, Saudi Arabia
| |
Collapse
|
31
|
Pollini T, Tran T, Wong P, Adam MA, Alseidi A, Corvera C, Hirose K, Nakakura E, Warren R, Maker VK, Maker AV. Improved survival of patients receiving immunotherapy and chemotherapy following curative-intent resection of colorectal liver metastases. J Gastrointest Surg 2024; 28:246-251. [PMID: 38445916 DOI: 10.1016/j.gassur.2023.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/25/2023] [Indexed: 03/07/2024]
Abstract
BACKGROUND Despite significant advancements in the treatment of patients with colorectal liver metastases (CRLMs), only a minority will experience long-term survival. This study aimed to determine the effect of chemotherapy (CT) and immunotherapy (IT) compared with that of CT alone on patient survival after surgical resection. METHODS Patients undergoing curative-intent liver resection followed by adjuvant systemic therapy for stage IV colon cancer were identified using the National Cancer Database. Patients were stratified into type of therapy (CT alone vs CT + IT) and microsatellite status. Propensity score-weighted analysis was performed through 1:1 matching based on the nearest neighbor method. RESULTS Of 9943 patients who underwent resection of CRLMs, 7971 (80%) received systemic adjuvant therapy. Of 7971 patients, 1432 (18%) received a combination of CT and IT. Microsatellite status was not associated with overall survival (OS). Adjuvant CT + IT was associated with increased 3-year OS compared with that of CT alone in both the unmatched cohort (55% vs 48%, respectively; P < .001) and matched cohort (52% vs 48%, respectively; P = .050). On multivariate analysis, older age, positive resection margins, and KRAS mutation were independent predictors of poor survival, whereas the administration of adjuvant CT + IT was an independent predictor of improved survival. CONCLUSION IT combined with CT was associated with improved survival compared with that of CT alone after curative-intent resection of CRLMs, regardless of microsatellite instability status. Clinical trials to determine optimal patient selection, IT regimen, and long-term efficacy to improve outcomes of patients with CRLMs are warranted.
Collapse
Affiliation(s)
- Tommaso Pollini
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Thuy Tran
- Division of Surgical Oncology, Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte, California, United States; Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Paul Wong
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Mohamed A Adam
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Adnan Alseidi
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Carlos Corvera
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Kenzo Hirose
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Eric Nakakura
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Robert Warren
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Vijay K Maker
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of California San Francisco, San Francisco, California, United States.
| |
Collapse
|
32
|
Ciepiela I, Szczepaniak M, Ciepiela P, Hińcza-Nowak K, Kopczyński J, Macek P, Kubicka K, Chrapek M, Tyka M, Góźdź S, Kowalik A. Tumor location matters, next generation sequencing mutation profiling of left-sided, rectal, and right-sided colorectal tumors in 552 patients. Sci Rep 2024; 14:4619. [PMID: 38409377 PMCID: PMC10897470 DOI: 10.1038/s41598-024-55139-w] [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: 04/30/2023] [Accepted: 02/20/2024] [Indexed: 02/28/2024] Open
Abstract
Despite the introduction of new molecular classifications, advanced colorectal cancer (CRC) is treated with chemotherapy supplemented with anti-EGFR and anti-VEGF targeted therapy. In this study, 552 CRC cases with different primary tumor locations (250 left side, 190 rectum, and 112 right side) were retrospectively analyzed by next generation sequencing for mutations in 50 genes. The most frequently mutated genes were TP53 in left-sided tumors compared to right-sided tumors and BRAF in right-sided tumors compared to left-sided tumors. Mutations in KRAS, NRAS, and BRAF were not detected in 45% of patients with left-sided tumors and in 28.6% of patients with right-sided tumors. Liver metastases were more common in patients with left-sided tumors. Tumors on the right side were larger at diagnosis and had a higher grade (G3) than tumors on the left. Rectal tumors exhibit distinctive biological characteristics when compared to left-sided tumors, including a higher absence rate of KRAS, NRAS, and BRAF mutations (47.4% in rectal versus 42.8% in left-sided tumors). These rectal tumors are also unique in their primary metastasis site, which is predominantly the lungs, and they have varying mutation rates, particularly in genes such as BRAF, FBXW7, and TP53, that distinguish them from tumors found in other locations. Primary tumor location has implications for the potential treatment of CRC with anti-EGFR therapy.
Collapse
Affiliation(s)
- Izabela Ciepiela
- Radiotherapy Department, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Magdalena Szczepaniak
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Przemysław Ciepiela
- Surgical Oncology Department, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Kinga Hińcza-Nowak
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, 25-734, Kielce, Poland
- Endocrinology Clinic, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Janusz Kopczyński
- Surgical Pathology, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Paweł Macek
- Collegium Medicum, Jan Kochanowski University, 25-319, Kielce, Poland
- Department of Epidemiology and Cancer Control, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Kamila Kubicka
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Magdalena Chrapek
- Department of Mathematics, Faculty of Natural Sciences, Jan Kochanowski University, 25-406, Kielce, Poland
| | - Magdalena Tyka
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Stanisław Góźdź
- Collegium Medicum, Jan Kochanowski University, 25-319, Kielce, Poland
- Clinical Oncology Clinic, Holy Cross Cancer Centre, 25-734, Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, 25-734, Kielce, Poland.
- Division of Medical Biology, Institute of Biology, Jan Kochanowski University, 25-406, Kielce, Poland.
| |
Collapse
|
33
|
Mohite P, Yadav V, Pandhare R, Maitra S, Saleh FM, Saleem RM, Al-malky HS, Kumarasamy V, Subramaniyan V, Abdel-Daim MM, Uti DE. Revolutionizing Cancer Treatment: Unleashing the Power of Viral Vaccines, Monoclonal Antibodies, and Proteolysis-Targeting Chimeras in the New Era of Immunotherapy. ACS OMEGA 2024; 9:7277-7295. [PMID: 38405458 PMCID: PMC10882662 DOI: 10.1021/acsomega.3c06501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 02/27/2024]
Abstract
In the realm of cancer immunotherapy, a profound evolution has ushered in sophisticated strategies that encompass both traditional cancer vaccines and emerging viral vaccines. This comprehensive Review offers an in-depth exploration of the methodologies, clinical applications, success stories, and future prospects of these approaches. Traditional cancer vaccines have undergone significant advancements utilizing diverse modalities such as proteins, peptides, and dendritic cells. More recent innovations have focused on the physiological mechanisms enabling the human body to recognize and combat precancerous and malignant cells, introducing specific markers like peptide-based anticancer vaccines targeting tumor-associated antigens. Moreover, cancer viral vaccines, leveraging engineered viruses to stimulate immune responses against specific antigens, exhibit substantial promise in inducing robust and enduring immunity. Integration with complementary therapeutic methods, including monoclonal antibodies, adjuvants, and radiation therapy, has not only improved survival rates but also deepened our understanding of viral virulence. Recent strides in vaccine design, encompassing oncolytic viruses, virus-like particles, and viral vectors, mark the frontier of innovation. While these advances hold immense potential, critical challenges must be addressed, such as strategies for immune evasion, potential off-target effects, and the optimization of viral genomes. In the landscape of immunotherapy, noteworthy innovations take the spotlight from the use of immunomodulatory agents for the enhancement of innate and adaptive immune collaboration. The emergence of proteolysis-targeting chimeras (PROTACs) as precision tools for cancer therapy is particularly exciting. With a focus on various cancers, from melanoma to formidable solid tumors, this Review critically assesses types of cancer vaccines, mechanisms, barriers in vaccine therapy, vaccine efficacy, safety profiles, and immune-related adverse events, providing a nuanced perspective on the underlying mechanisms involving cytotoxic T cells, natural killer cells, and dendritic cells. The Review also underscores the transformative potential of cutting-edge technologies such as clinical studies, molecular sequencing, and artificial intelligence in advancing the field of cancer vaccines. These tools not only expedite progress but also emphasize the multidimensional and rapidly evolving nature of this research, affirming its profound significance in the broader context of cancer therapy.
Collapse
Affiliation(s)
- Popat Mohite
- AETs
St. John Institute of Pharmacy and Research, Palghar, Maharashtra 401404, India
| | - Vaishnavi Yadav
- AETs
St. John Institute of Pharmacy and Research, Palghar, Maharashtra 401404, India
| | - Ramdas Pandhare
- MESs
College of Pharmacy, Sonai Tal-Newasa, Maharashtra 414105, India
| | - Swastika Maitra
- Center
for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai 600077, India
- Department
of Microbiology, Adamas University, Kolkata 700 126, West Bengal, India
| | - Fayez M. Saleh
- Department
of Medical Microbiology, Faculty of Medicine, University of Tabuk, Tabuk 71491, Saudi Arabia
| | - Rasha Mohammed Saleem
- Department
of Laboratory Medicine, Faculty of Applied Medical Sciences, Al-Baha University, Al-Baha 65431, Saudi Arabia
| | - Hamdan S. Al-malky
- Regional
Drug Information Center, Ministry of Health, Jeddah 11176, Saudi Arabia
| | - Vinoth Kumarasamy
- Department
of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology
Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University, Jalan Lagoon Selatan, Bandar
Sunway, 47500 Selangor
Darul Ehsan, Malaysia
- Center
for Transdisciplinary Research, Department of Pharmacology, Savetha
Dental College, Savetha Institute of Medical and Technical Sciences, Savetha University, Chennai, Tamil Nadu 600077, India
| | - Mohamed M. Abdel-Daim
- Department
of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box
6231, Jeddah 21442, Saudi Arabia
- Pharmacology
Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Daniel E. Uti
- Department
of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine, Federal University of Health Sciences, Otukpo, Benue State 970001, Nigeria
| |
Collapse
|
34
|
Mei S, Peng S, Vong EG, Zhan J. A dual-functional oncolytic adenovirus ZD55-aPD-L1 scFv armed with PD-L1 inhibitor potentiates its antitumor activity. Int Immunopharmacol 2024; 128:111579. [PMID: 38278066 DOI: 10.1016/j.intimp.2024.111579] [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: 11/28/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Clinical data indicate that a substantial portion of cancer patients, though eligible for immune checkpoint inhibitor (ICI) therapy, cannot fully benefit from ICI monotherapy due to the poor immunogenicity of tumors and lack of tumor-infiltrating lymphocytes within the 'cold' tumor microenvironment (TME). In addition to poor antibody penetrance into the TME, systemic delivery of ICIs is associated with immune-related adverse events (irAEs) among recipients, some of which are life-threatening. Oncolytic virotherapy is a potentially viable approach to improving the efficacy of ICI therapy because of their ability to selectively replicate and lyse tumor cells, release tumor-associated antigens (TAAs), induce inflammatory response and promote lymphocyte infiltration in tumors. METHODS A recombinant oncolytic adenoviruses (OAd), denoted ZD55-aPD-L1 scFv, which carried the expression cassette for anti-PD-L1 scFv was constructed by molecular cloning. Western blot and ELISA assay were performed to detect aPD-L1 scFv expression. Flow cytometry were used to analyse PD-L1 expression and count tumor cells. Co-culture assay of human peripheral blood mononuclear cells (PBMCs) with tumor cells in vitro and triple-negative breast cancer (TNBC) MDA-MB-231 tumor-bearing model in vivo were evaluated the antitumor effects of recombinant oncolytic adenoviruses ZD55-aPD-L1 scFv. RESULTS We found that cells infected with recombinant oncolytic adenovirus ZD55-aPD-L1 scFv can effectively express aPD-L1 scFv, which function similarly to its full-length anti-PD-L1 antibody. PBMCs have inherently very limited killing effect on tumor cells even with administration of anti-PD-L1 antibody as observed from our in vitro co-cultures. Treatment consisting of ZD55 alone or ZD55 combined with anti-PD-L1 antibody yielded mediocre antitumor efficacy in subsequent in vitro and in vivo investigations, but were all substantially surpassed by the synergistic antitumor effects observed with ZD55-aPD-L1 scFv treatment. We show that the concomitant direct oncolysis by the recombinant OAd and localized autocrine/paracrine interception of PD-1:PD-L1 checkpoint interaction mediated by ZD55-aPD-L1 scFv-infected cells is exceedingly superior to co-administration of ZD55 and anti-PD-L1 antibody in the human TNBC mice model. CONCLUSIONS Our results provided evidence for the development of novel strategies, in this case an anti-PD-L1 scFv-armed OAd, to bolster immune responses to 'cold' tumors and to improve therapeutic responsiveness to ICIs.
Collapse
Affiliation(s)
- Shengsheng Mei
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, China
| | - Shanshan Peng
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, China
| | - Eu Gene Vong
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, China
| | - Jinbiao Zhan
- Department of Biochemistry, Cancer Institute of the Second Affiliated Hospital (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education), School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
35
|
Song Y, Mao Q, Zhou M, Liu CJ, Kong L, Hu T. Effectiveness of bevacizumab in the treatment of metastatic colorectal cancer: a systematic review and meta-analysis. BMC Gastroenterol 2024; 24:58. [PMID: 38302922 PMCID: PMC10832121 DOI: 10.1186/s12876-024-03134-w] [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: 05/09/2023] [Accepted: 01/14/2024] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVE To evaluate the benefit of bevacizumab under the comprehensive treatment strategy and its advantages over other drugs, so as to provide reference for the formulation of clinical plans. METHODS As of October 1, 2022, the randomized controlled clinical trials of bevacizumab in combination with metastatic colorectal cancer published in PubMed, Cochrane Library and Medline databases were searched. The odds ratio (OR) and its 95% confidence interval (CI) were used to evaluate the short-term disease control effect and long-term survival of the treatment strategy. RESULTS 21 RCTs (6665 patients; 3356 patients in the experimental group and 3309 patients in the control group; average age, 55-75 years) were treated with bevacizumab as the experimental group for metastatic colorectal cancer. BEV has stronger anti-tumor activity than the single treatment scheme (OR = 1.30, 95% CI: 1.11-1.52). And Benefits of the BEV group were 0.73 (0.55, 0.96), 1.26 (0.71, 2.24), 1.63 (0.92, 2.87) and 0.07 (0.02, 0.25) compared with CET, VAN, CED and PAN respectively. The disease control of BEV combined therapy was better (OR = 1.36, 95% CI: 1.04-1.78). The same as compared with cediranib (OR = 1.94, 95% CI: 1.06-3.55). However, the long-term prognosis of BEV, including the overall survival (HRs = 0.98, 95% CI: 0.84-1.15) and progression-free survival (HRs = 1.05,95% CI: 0.97-1.13) were not prolonged. The survival benefits of cetuximab and panitumumab were not reflected. CONCLUSION The addition of BEV can enhance the anti-tumor ability and disease control, while cetuximab and panitumumab may have stronger ability. However, it did not effectively improve the survival of patients. A more reasonable and effective treatment plan needs more clinical experimental support.
Collapse
Affiliation(s)
- Yu Song
- Department of Intensive Care Unit, the First Affiliated Hospital of Shandong Traditional Medical University, 250000, Jinan, China
| | - Qianqian Mao
- Department of Oncology, School of Medicine, Zhongda Hospital, Southeast University, No. 87 Dingjiaqiao Road, 210009, Nanjing, Jiangsu Province, China
| | - Manling Zhou
- Department of Oncology, Shenzhen Longhua District Central Hospital, Shenzhen, China
| | - Cheng-Jiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, 246000, Anqing, AnHui, China.
| | - Li Kong
- Department of Intensive Care Unit, the First Affiliated Hospital of Shandong Traditional Medical University, 250000, Jinan, China.
| | - Ting Hu
- Department of General Practice, Anqing Municipal Hospital, 246000, Anqing, AnHui, China
| |
Collapse
|
36
|
Wu Z, Zhou S, Liang D, Mu L. GPX2 acts as an oncogene and cudraflavone C has an anti-tumor effect by suppressing GPX2-dependent Wnt/β-catenin pathway in colorectal cancer cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1115-1125. [PMID: 37610461 DOI: 10.1007/s00210-023-02668-2] [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: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023]
Abstract
Colorectal carcinoma (CRC) is a common cancer associated with poor prognosis, and cudraflavone C (Cud C) is a natural flavonol with reported anti-CRC capacity. However, the precise mechanisms underlying the anti-CRC effect require further demonstration. The aim of present study was to evaluate the impact of Cud C on the cell viability and apoptosis of CRC cells and to determine the underlying mechanisms. The Human Protein Atlas (THPA) and Gene Expression Profiling Interactive Analysis (GEPIA) databases were used to analyze the expression status of glutathione peroxidase 2 (GPX2) in CRC. Cell viability was examined using cell counting kit-8 (CCK-8) assay. Flow cytometry was utilized to evaluate apoptosis. The levels of gene transcription and protein expression of GPX2, caspase-3, cleaved caspase-3), β-catenin, and c-Myc were determined by RT-qPCR and Western blotting. Our results showed that GPX2 was overexpressed in CRC as compared to normal tissue and the extent of GPX2 overexpression is greatest in CRC when compared with other cancers according to GEPIA and THPA databases. GPX2 knockdown significantly suppressed the cell viability, induced apoptosis of CRC cell lines, and restrained the activity of Wnt/β-catenin pathway. Cud C treatment decreased cell viability, induced apoptosis in CRC cell lines, and diminished the expression level of GPX2-dependent activation of Wnt/β-catenin pathway, while such effects can be abolished by GPX2 overexpression. In conclusion, Cud C suppressed GPX2-dependent Wnt/β-catenin pathway to exert anti-CRC function.
Collapse
Affiliation(s)
- Zhuo Wu
- Uutpatient Department, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Su Zhou
- Department of Drug Management, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, People's Republic of China
| | - Dan Liang
- Department of Otolaryngology, the First Affiliated Hospital of Jinzhou Medical University, 5-2 Renmin Street, Jinzhou, People's Republic of China
| | - Lan Mu
- Department of Otolaryngology, the First Affiliated Hospital of Jinzhou Medical University, 5-2 Renmin Street, Jinzhou, People's Republic of China.
| |
Collapse
|
37
|
Ahluwalia P, Ballur K, Leeman T, Vashisht A, Singh H, Omar N, Mondal AK, Vaibhav K, Baban B, Kolhe R. Incorporating Novel Technologies in Precision Oncology for Colorectal Cancer: Advancing Personalized Medicine. Cancers (Basel) 2024; 16:480. [PMID: 38339232 PMCID: PMC10854941 DOI: 10.3390/cancers16030480] [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: 10/31/2023] [Revised: 01/10/2024] [Accepted: 01/13/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most heterogeneous and deadly diseases, with a global incidence of 1.5 million cases per year. Genomics has revolutionized the clinical management of CRC by enabling comprehensive molecular profiling of cancer. However, a deeper understanding of the molecular factors is needed to identify new prognostic and predictive markers that can assist in designing more effective therapeutic regimens for the improved management of CRC. Recent breakthroughs in single-cell analysis have identified new cell subtypes that play a critical role in tumor progression and could serve as potential therapeutic targets. Spatial analysis of the transcriptome and proteome holds the key to unlocking pathogenic cellular interactions, while liquid biopsy profiling of molecular variables from serum holds great potential for monitoring therapy resistance. Furthermore, gene expression signatures from various pathways have emerged as promising prognostic indicators in colorectal cancer and have the potential to enhance the development of equitable medicine. The advancement of these technologies for identifying new markers, particularly in the domain of predictive and personalized medicine, has the potential to improve the management of patients with CRC. Further investigations utilizing similar methods could uncover molecular subtypes specific to emerging therapies, potentially strengthening the development of personalized medicine for CRC patients.
Collapse
Affiliation(s)
- Pankaj Ahluwalia
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kalyani Ballur
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Tiffanie Leeman
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashutosh Vashisht
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Harmanpreet Singh
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Nivin Omar
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Ashis K. Mondal
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| | - Kumar Vaibhav
- Department of Neurosurgery, Augusta University, Augusta, GA 30912, USA;
| | - Babak Baban
- Departments of Neurology and Surgery, Augusta University, Augusta, GA 30912, USA;
| | - Ravindra Kolhe
- Department of Pathology, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA; (P.A.); (K.B.); (T.L.); (A.V.); (H.S.); (N.O.); (A.K.M.)
| |
Collapse
|
38
|
Arroyo-Olarte R, Mejía-Muñoz A, León-Cabrera S. Expanded Alternatives of CRISPR-Cas9 Applications in Immunotherapy of Colorectal Cancer. Mol Diagn Ther 2024; 28:69-86. [PMID: 37907826 PMCID: PMC10786962 DOI: 10.1007/s40291-023-00680-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2023] [Indexed: 11/02/2023]
Abstract
Immunotherapy for colorectal cancer (CRC) is limited to patients with advanced disease who have already undergone first-line chemotherapy and whose tumors exhibit microsatellite instability. Novel technical strategies are required to enhance therapeutic options and achieve a more robust immunological response. Therefore, exploring gene analysis and manipulation at the molecular level can further accelerate the development of advanced technologies to address these challenges. The emergence of advanced genome editing technology, particularly of clustered, regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein (Cas) 9, holds promise in expanding the boundaries of cancer immunotherapy. In this manuscript, we provide a comprehensive review of the applications and perspectives of CRISPR technology in improving the design, generation, and efficiency of current immunotherapies, focusing on solid tumors such as colorectal cancer, where these approaches have not been as successful as in hematological conditions.
Collapse
Affiliation(s)
- Rubén Arroyo-Olarte
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, 54090, Tlalnepantla, Edo. De México, México
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, 54090, Tlalnepantla, Edo. De México, México
| | - Aranza Mejía-Muñoz
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, 54090, Tlalnepantla, Edo. De México, México
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, 54090, Tlalnepantla, Edo. De México, México
| | - Sonia León-Cabrera
- Unidad de Biomedicina, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, 54090, Tlalnepantla, Edo. De México, México.
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, 54090, Tlalnepantla, Edo. De México, México.
| |
Collapse
|
39
|
Gawesh ZM, Ibrahim EM, ElKalla HMHR, Awad AAH, Mohamed MA. Evaluation of HHLA2 and CD8 Immunohistochemical Expression in Colorectal Carcinoma and Their Prognostic Significance. Asian Pac J Cancer Prev 2023; 24:4309-4319. [PMID: 38156868 PMCID: PMC10909113 DOI: 10.31557/apjcp.2023.24.12.4309] [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: 08/27/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Colorectal carcinoma (CRC) is the third most common malignancy worldwide. Human endogenous retrovirus H long terminal repeat-associating protein 2 (HHLA2) is a novel immune checkpoint molecule. The association between HHLA2 expression and clinicopathological features and its prognostic significance in CRC patients are still controversial. The aim of this study is to evaluate the prognostic value of immunohistochemical (IHC) expression of HHLA2 and CD8 in CRC. MATERIAL AND METHODS This retrospective study included 134 cases diagnosed with primary CRC at the Gastrointestinal Surgery Center (GISC) department, Mansoura Faculty of Medicine, during the period from December 2014 to December 2018. Clinicopathological and survival data were collected. IHC for HHLA2 and CD8 was performed, and they were correlated with clinicopathological parameters and patient prognosis. RESULTS Among 134 CRC cases, high HHLA2 expression was detected in 73 (54.5%). High HHLA2 expression was significantly related to the depth of invasion (P = 0.005*), lymph node metastasis (P = 0.01*), tumor stage )P = 0.002*), and distant recurrence )P = 0.012*). Multivariate analysis spotted HHLA2 high expression as an independent prognostic predictor for OS in CRC (P = 0.03*) and DFS (P = 0.008*). CD8 shows a significant correlation with tumor infiltrating lymphocytes (TILs) (P ≤ 0.001*), absence of metastasis ((P = 0.029*), absence of tumor deposits (P=0.014*). However, CD8 shows no significant association with survival or HHLA2. CONCLUSION HHLA2 is an independent prognostic factor for the overall survival and disease free survival of CRC patients and can predict poor prognosis in CRC patients.
Collapse
Affiliation(s)
| | | | | | | | - Mie Ali Mohamed
- Department of Pathology, Mansoura Faculty of Medicine, Mansoura, Egypt.
| |
Collapse
|
40
|
Das S, Acharya D. Immunological Assessment of Recent Immunotherapy for Colorectal Cancer. Immunol Invest 2023; 52:1065-1095. [PMID: 37812224 DOI: 10.1080/08820139.2023.2264906] [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: 10/10/2023]
Abstract
Colorectal cancer (CRC) is the third most prevalent malignancy with increased incidence and mortality rates worldwide. Traditional treatment approaches have attempted to efficiently target CRC; however, they have failed in most cases, owing to the cytotoxicity and non-specificity of these therapies. Therefore, it is essential to develop an effective alternative therapy to improve the clinical outcomes in heterogeneous CRC cases. Immunotherapy has transformed cancer treatment with remarkable efficacy and overcomes the limitations of traditional treatments. With an understanding of the cancer-immunity cycle and tumor microenvironment evolution, current immunotherapy approaches have elicited enhanced antitumor immune responses. In this comprehensive review, we outline the latest advances in immunotherapy targeting CRC and provide insights into antitumor immune responses reported in landmark clinical studies. We focused on highlighting the combination approaches that synergistically induce immune responses and eliminate immunosuppression. This review aimed to understand the limitations and potential of recent immunotherapy clinical studies conducted in the last five years (2019-2023) and to transform this knowledge into a rational design of clinical trials intended for effective antitumor immune responses in CRC.
Collapse
Affiliation(s)
- Subhadeep Das
- Department of Biotechnology, GIET University, Gunupur, India
| | | |
Collapse
|
41
|
Chang L, Jung NY, Atari A, Rodriguez DJ, Kesar D, Song TY, Rees MG, Ronan M, Li R, Ruiz P, Chaturantabut S, Ito T, van Tienen LM, Tseng YY, Roth JA, Sellers WR. Systematic profiling of conditional pathway activation identifies context-dependent synthetic lethalities. Nat Genet 2023; 55:1709-1720. [PMID: 37749246 DOI: 10.1038/s41588-023-01515-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 08/22/2023] [Indexed: 09/27/2023]
Abstract
The paradigm of cancer-targeted therapies has focused largely on inhibition of critical pathways in cancer. Conversely, conditional activation of signaling pathways as a new source of selective cancer vulnerabilities has not been deeply characterized. In this study, we sought to systematically identify context-specific gene-activation-induced lethalities in cancer. To this end, we developed a method for gain-of-function genetic perturbations simultaneously across ~500 barcoded cancer cell lines. Using this approach, we queried the pan-cancer vulnerability landscape upon activating ten key pathway nodes, revealing selective activation dependencies of MAPK and PI3K pathways associated with specific biomarkers. Notably, we discovered new pathway hyperactivation dependencies in subsets of APC-mutant colorectal cancers where further activation of the WNT pathway by APC knockdown or direct β-catenin overexpression led to robust antitumor effects in xenograft and patient-derived organoid models. Together, this study reveals a new class of conditional gene-activation dependencies in cancer.
Collapse
Affiliation(s)
- Liang Chang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nancy Y Jung
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Adel Atari
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Devishi Kesar
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tian-Yu Song
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Melissa Ronan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ruitong Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paloma Ruiz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Saireudee Chaturantabut
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Biopharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Takahiro Ito
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Laurens M van Tienen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yuen-Yi Tseng
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - William R Sellers
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Akce M, Farran B, Switchenko JM, Rupji M, Kang S, Khalil L, Ruggieri-Joyce A, Olson B, Shaib WL, Wu C, Alese OB, Diab M, Lesinski GB, El-Rayes BF. Phase II trial of nivolumab and metformin in patients with treatment-refractory microsatellite stable metastatic colorectal cancer. J Immunother Cancer 2023; 11:e007235. [PMID: 37852737 PMCID: PMC10603338 DOI: 10.1136/jitc-2023-007235] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND Preclinical studies showed metformin reduces exhaustion of tumor-infiltrating lymphocytes and potentiates programmed cell death protein-1 (PD-1) blockade. We hypothesized that metformin with nivolumab would elicit potent antitumor and immune modulatory activity in metastatic microsatellite stable (MSS) colorectal cancer (CRC). We evaluated this hypothesis in a phase II study. METHODS Nivolumab (480 mg) was administered intravenously every 4 weeks while metformin (1000 mg) was given orally, two times per day following a 14-day metformin only lead-in phase. Patients ≥18 years of age, with previously treated, stage IV MSS CRC, and Eastern Cooperative Oncology Group 0-1, having received no prior anti-PD-1 agent were eligible. The primary endpoint was overall response rate with secondary endpoints of overall survival (OS) and progression-free survival (PFS). Correlative studies using paired pretreatment/on-treatment biopsies and peripheral blood evaluated a series of immune biomarkers in the tumor microenvironment and systemic circulation using ChipCytometry and flow cytometry. RESULTS A total of 24 patients were enrolled, 6 patients were replaced per protocol, 18 patients had evaluable disease. Of the 18 evaluable patients, 11/18 (61%) were women and the median age was 58 (IQR 50-67). Two patients had stable disease, but no patients had objective response, hence the study was stopped for futility. Median OS and PFS was 5.2 months (95% CI (3.2 to 11.7)) and 2.3 months (95% CI (1.7 to 2.3)). Most common grade 3/4 toxicities: Anemia (n=2), diarrhea (n=2), and fever (n=2). Metformin alone failed to increase the infiltration of T-cell subsets in the tumor, but combined metformin and nivolumab increased percentages of tumor-infiltrating leukocytes (p=0.031). Dual treatment also increased Tim3+ levels in patient tissues and decreased naïve CD8+T cells (p=0.0475). CONCLUSIONS Nivolumab and metformin were well tolerated in patients with MSS CRC but had no evidence of efficacy. Correlative studies did not reveal an appreciable degree of immune modulation from metformin alone, but showed trends in tumorous T-cell infiltration as a result of dual metformin and PD-1 blockade despite progression in a majority of patients.
Collapse
Affiliation(s)
- Mehmet Akce
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Jeffrey M Switchenko
- Biostatistics Shared Resource, Emory University Winship Cancer institute, Atlanta, Georgia, USA
- Department of Biostsatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Manali Rupji
- Biostatistics Shared Resource, Emory University Winship Cancer institute, Atlanta, Georgia, USA
| | - Sandra Kang
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Lana Khalil
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Amanda Ruggieri-Joyce
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Brian Olson
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Walid L Shaib
- Northwest Georgia Oncology Centers Wellstar, Marietta, Georgia, USA
| | - Christina Wu
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Arizona, Scottsdale, Arizona, USA
| | - Olatunji B Alese
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Maria Diab
- Department of Internal Medicine, Michigan State University, East Lansing, Michigan, USA
- Department of Internal Medicine, Henry Ford Health, Detroit, Michigan, USA
| | - Gregory B Lesinski
- Department of Hematology and Medical Oncology, Emory University Winship Cancer Institute, Atlanta, Georgia, USA
| | - Bassel F El-Rayes
- Division of Hematology and Oncology, Department of Medicine, O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, USA
| |
Collapse
|
43
|
Lingas EC. Early-Onset Colon Cancer: A Narrative Review of Its Pathogenesis, Clinical Presentation, Treatment, and Prognosis. Cureus 2023; 15:e45404. [PMID: 37854763 PMCID: PMC10579844 DOI: 10.7759/cureus.45404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2023] [Indexed: 10/20/2023] Open
Abstract
Colon cancer remains a leading cause of cancer-related deaths, and there has been a rise in the incidence of early-onset colon cancer or colon cancer diagnosed before the age of 50 years old. Early-onset colon cancer has several differences in clinical presentation, as well as histopathology, genetic alteration, and molecular profiling. Early-onset colon cancer can be differentiated into familial type that includes hereditary familial syndrome and sporadic type. Demographic variance also exists in both developing and developed countries. Due to the rising incidence of colon cancer diagnosed in younger age, it is imperative to examine the available evidence regarding the mortality rate of early-onset colon cancer. Colon cancer is affected by numerous modifiable and non-modifiable risk factors. Increasing obesity and lifestyle disorders in the younger population, such as smoking, may influence this increasing trend. There are existing guidelines for colon cancer screening in both average-risk and high-risk individuals. This narrative review aims to highlight the pathogenesis of early-onset CRC; its clinical presentation, treatment, prognosis; and how it differs from late-onset CRC.
Collapse
Affiliation(s)
- Elvina C Lingas
- Hospital Medicine, New York University (NYU) Langone Health Long Island Community Hospital, Patchogue, USA
| |
Collapse
|
44
|
He Y, Hong C, Huang S, Kaskow JA, Covarrubias G, Pires IS, Sacane JC, Hammond PT, Belcher AM. STING Protein-Based In Situ Vaccine Synergizes CD4 + T, CD8 + T, and NK Cells for Tumor Eradication. Adv Healthc Mater 2023; 12:e2300688. [PMID: 37015729 PMCID: PMC10964211 DOI: 10.1002/adhm.202300688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/15/2023] [Indexed: 04/06/2023]
Abstract
Stimulator of interferon genes (STING) signaling is a promising target in cancer immunotherapy, with many ongoing clinical studies in combination with immune checkpoint blockade (ICB). Existing STING-based therapies largely focus on activating CD8+ T cell or NK cell-mediated cytotoxicity, while the role of CD4+ T cells in STING signaling has yet to be extensively studied in vivo. Here, a distinct CD4-mediated, protein-based combination therapy of STING and ICB as an in situ vaccine, is reported. The treatment eliminates subcutaneous MC38 and YUMM1.7 tumors in 70-100% of mice and protected all cured mice against rechallenge. Mechanistic studies reveal a robust TH 1 polarization and suppression of Treg of CD4+ T cells, followed by an effective collaboration of CD4+ T, CD8+ T, and NK cells to eliminate tumors. Finally, the potential to overcome host STING deficiency by significantly decreasing MC38 tumor burden in STING KO mice is demonstrated, addressing the translational challenge for the 19% of human population with loss-of-function STING variants.
Collapse
Affiliation(s)
- Yanpu He
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Celestine Hong
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Shengnan Huang
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Material Science and EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Justin A. Kaskow
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Gil Covarrubias
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Ivan S. Pires
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - James C. Sacane
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Paula T. Hammond
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Angela M. Belcher
- Koch Institute for Integrative Cancer ResearchMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
- Department of Material Science and EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| |
Collapse
|
45
|
Aljama S, Lago EP, Zafra O, Sierra J, Simón D, Santos C, Pascual JR, Garcia-Romero N. Dichotomous colorectal cancer behaviour. Crit Rev Oncol Hematol 2023; 189:104067. [PMID: 37454703 DOI: 10.1016/j.critrevonc.2023.104067] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common malignant tumor and one of the deadliest cancers. At molecular level, CRC is a heterogeneous disease that could be divided in four Consensus Molecular Subtypes. Given the differences in the disease due to its anatomical location (proximal and distal colon), another classification should be considered. Here, we review the current knowledge on CRC dichotomic´s behaviour based on two different entities; right and left-sided tumors, their impact on clinical trial data, microbiota spatial composition and the interaction with the nervous system. We discuss recent advances in understanding how the spatial tumor heterogeneity influences the tumor growth, progression, and responses to current therapies.
Collapse
Affiliation(s)
- Sara Aljama
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Estela P Lago
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Olga Zafra
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Javier Sierra
- Faculty of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Diana Simón
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | - Cruz Santos
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain
| | | | - Noemi Garcia-Romero
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria, 28223 Madrid, Spain.
| |
Collapse
|
46
|
Alrosan AZ, Alrosan K, Heilat GB, Alsharedeh R, Abudalo R, Oqal M, Alqudah A, Elmaghrabi YA. Potential roles of NEDD4 and NEDD4L and their utility as therapeutic targets in high‑incidence adult male cancers (Review). Mol Clin Oncol 2023; 19:68. [PMID: 37614371 PMCID: PMC10442760 DOI: 10.3892/mco.2023.2664] [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: 02/18/2023] [Accepted: 07/07/2023] [Indexed: 08/25/2023] Open
Abstract
The term 'cancer' refers to >100 disorders that progressively manifest over time and are characterized by uncontrolled cell division. Although malignant growth can occur in virtually any human tissue, the underlying mechanisms underlying all forms of cancer are consistent. The International Agency for Research on Cancer's annual GLOBOCAN 2020 report provided an update on the global cancer incidence and mortality. Excluding non-melanoma skin cancer, the report predicts that there will be 19.3 million new cancer cases and >10 million cancer-related fatalities in 2023. Lung, prostate, and colon cancers are the most prevalent and lethal cancers in males. It was recognized that post-translational modifications (PTMs) of proteins are necessary for almost all cellular biological processes, as well as in cancer development and metastasis to other bodily organs. Thus, PTMs have a considerable impact on how proteins behave. Various PTMs may have harmful roles by affecting the hallmarks of cancer, metabolism and the regulation of the tumor microenvironment. PTMs and genetic changes/mutations are essential in carcinogenesis and cancer development. A pivotal PTM mechanism is protein ubiquitination. Of note, the rate-limiting stage of the protein ubiquitination cascade is hypothesized to be E3-ligase-mediated ubiquitination. Numerous studies revealed that the neural precursor cell expressed developmentally downregulated protein 4 (NEDD4) E3 ligase is among the E3 ubiquitin ligases that have essential roles in cellular processes. It regulates protein degradation and substrate ubiquitination. In addition, it has been shown that NEDD4 primarily functions as an oncogene in various malignancies but can also act as a tumor suppressor in certain types of tumor. In the present review, the roles of NEDD4 as an anticancer protein in various high-incidence male malignancies and the significance of NEDD4 as a potential cancer therapeutic target are discussed. In addition, the targeting of NEDD4 as a therapeutic strategy for the treatment of human malignancies is explored.
Collapse
Affiliation(s)
- Amjad Z. Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Khaled Alrosan
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Ghaith B. Heilat
- Department of General Surgery and Urology, Faculty of Medicine, The Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Rawan Alsharedeh
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The Yarmouk University, Irbid 21163, Jordan
| | - Rawan Abudalo
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | - Muna Oqal
- Department of Pharmaceutical Technology, Faculty of Pharmacy, The Hashemite University, Zarqa 13133, Jordan
| | - Abdelrahim Alqudah
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, The Hashemite University, Zarqa 13133, Jordan
| | | |
Collapse
|
47
|
Guo XW, Lei RE, Zhou QN, Zhang G, Hu BL, Liang YX. Tumor microenvironment characterization in colorectal cancer to identify prognostic and immunotherapy genes signature. BMC Cancer 2023; 23:773. [PMID: 37596528 PMCID: PMC10436413 DOI: 10.1186/s12885-023-11277-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/08/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a crucial role in tumorigenesis, progression, and therapeutic response in many cancers. This study aimed to comprehensively investigate the role of TME in colorectal cancer (CRC) by generating a TMEscore based on gene expression. METHODS The TME patterns of CRC datasets were investigated, and the TMEscores were calculated. An unsupervised clustering method was used to divide samples into clusters. The associations between TMEscores and clinical features, prognosis, immune score, gene mutations, and immune checkpoint inhibitors were analyzed. A TME signature was constructed using the TMEscore-related genes. The results were validated using external and clinical cohorts. RESULTS The TME pattern landscape was for CRC was examined using 960 samples, and then the TMEscore pattern of CRC datasets was evaluated. Two TMEscore clusters were identified, and the high TMEscore cluster was associated with early-stage CRC and better prognosis in patients with CRC when compared with the low TMEscore clusters. The high TMEscore cluster indicated elevated tumor cell scores and tumor gene mutation burden, and decreased tumor purity, when compared with the low TMEscore cluster. Patients with high TMEscore were more likely to respond to immune checkpoint therapy than those with low TMEscore. A TME signature was constructed using the TMEscore-related genes superimposing the results of two machine learning methods (LASSO and XGBoost algorithms), and a TMEscore-related four-gene signature was established, which had a high predictive value for discriminating patients from different TMEscore clusters. The prognostic value of the TMEscore was validated in two independent cohorts, and the expression of TME signature genes was verified in four external cohorts and clinical samples. CONCLUSION Our study provides a comprehensive description of TME characteristics in CRC and demonstrates that the TMEscore is a reliable prognostic biomarker and predictive indicator for patients with CRC undergoing immunotherapy.
Collapse
Affiliation(s)
- Xian-Wen Guo
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Tao-Yuan Road, Nanning, 530021, Guangxi, China
| | - Rong-E Lei
- Department of Gastroenterology, First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Qing-Nan Zhou
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Tao-Yuan Road, Nanning, 530021, Guangxi, China
| | - Guo Zhang
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Tao-Yuan Road, Nanning, 530021, Guangxi, China
| | - Bang-Li Hu
- Department of Research, Guangxi Medical University Cancer Hospital, No.71 Hedi Road, Nanning, 530021, Guangxi, China.
| | - Yun-Xiao Liang
- Department of Gastroenterology, The People's Hospital of Guangxi Zhuang Autonomous Region, No.6 Tao-Yuan Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
48
|
Okoye C, Tran M, Soladoye E, Akahara DE, Emeasoba CM, Ojinna BT, Anasonye E, Obadare OO, Diala CS, Salaudeen BH, Evbayekha EO, Okobi OE. A Review of 10-Year Survivability of Immunotherapy in the Management of Colon Cancer. Cureus 2023; 15:e43189. [PMID: 37692610 PMCID: PMC10485874 DOI: 10.7759/cureus.43189] [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: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/12/2023] Open
Abstract
Colon cancer is one of the most common cancers in the United States of America. In addition to conventional treatment approaches such as surgery, chemotherapy, and radiation for colorectal cancer, immunotherapy has gained recognition over the past few years. However, its effectiveness in colorectal cancer treatment is controversial. Our study investigates the survival and progression-free rates of immunotherapy for different types of colorectal cancer over the last 10 years. We conducted literature reviews from various clinical trials and research studies to evaluate immunotherapy's role in colorectal cancer treatment. We also investigated how it affects clinical outcomes. We discovered a range of effective immunotherapy approaches targeting various growth factors and signaling pathways. These modalities include monoclonal antibodies aimed at growth factors such as epidermal growth factor (EGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), human epidermal growth factor receptor 2 (HER2), and downstream signaling pathways such as mitogen-activated protein kinase (MAPK), kirsten rat sarcoma viral oncogene (KRAS), B-raf proto-oncogene, serine/threonine kinase (BRAF), and phosphatase and tensin homolog (PTEN). Additionally, we identified immune checkpoint inhibitors, such as cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) inhibitors and programmed cell death ligand 1 (PD-L1) inhibitors, as well as target therapy and adoptive cell therapy as promising immunotherapeutic options. Nevertheless, the application of immunotherapy remains highly limited due to various factors influencing survival and progression-free rates, including tumor microenvironment, microsatellite instability, immune checkpoint expression, and gut microbiome. Additionally, its effectiveness is restricted to a small subgroup of patients, accompanied by side effects and the development of drug resistance mechanisms. To unlock its full potential, further clinical trials and research on molecular pathways in colorectal cancer are imperative. This will ultimately enhance drug discovery success and lead to more effective clinical management approaches.
Collapse
Affiliation(s)
- Chiugo Okoye
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - My Tran
- Internal Medicine, Baptist Health-University of Arkansas for Medical Sciences - Arkansas, North Little Rock, USA
| | | | | | | | - Blessing T Ojinna
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | | | | | - Chiamaka S Diala
- Health/Biomedical Informatics, The University of Texas Health Science Center, Houston, USA
- Internal Medicine, Piedmont Athens Regional, Athens, USA
| | | | | | - Okelue E Okobi
- Family Medicine, Medficient Health Systems, Laurel, USA
- Family Medicine, Lakeside Medical Center, Belle Glade, USA
| |
Collapse
|
49
|
Nelson VK, Nuli MV, Mastanaiah J, Saleem T. S. M, Birudala G, Jamous YF, Alshargi O, Kotha KK, Sudhan HH, Mani RR, Muthumanickam A, Niranjan D, Jain NK, Agrawal A, Jadon AS, Mayasa V, Jha NK, Kolesarova A, Slama P, Roychoudhury S. Reactive oxygen species mediated apoptotic death of colon cancer cells: therapeutic potential of plant derived alkaloids. Front Endocrinol (Lausanne) 2023; 14:1201198. [PMID: 37560308 PMCID: PMC10408138 DOI: 10.3389/fendo.2023.1201198] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 06/28/2023] [Indexed: 08/11/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most deaths causing diseases worldwide. Several risk factors including hormones like insulin and insulin like growth factors (e.g., IGF-1) have been considered responsible for growth and progression of colon cancer. Though there is a huge advancement in the available screening as well as treatment techniques for CRC. There is no significant decrease in the mortality of cancer patients. Moreover, the current treatment approaches for CRC are associated with serious challenges like drug resistance and cancer re-growth. Given the severity of the disease, there is an urgent need for novel therapeutic agents with ideal characteristics. Several pieces of evidence suggested that natural products, specifically medicinal plants, and derived phytochemicals may serve as potential sources for novel drug discovery for various diseases including cancer. On the other hand, cancer cells like colon cancer require a high basal level of reactive oxygen species (ROS) to maintain its own cellular functions. However, excess production of intracellular ROS leads to cancer cell death via disturbing cellular redox homeostasis. Therefore, medicinal plants and derived phytocompounds that can enhance the intracellular ROS and induce apoptotic cell death in cancer cells via modulating various molecular targets including IGF-1 could be potential therapeutic agents. Alkaloids form a major class of such phytoconstituents that can play a key role in cancer prevention. Moreover, several preclinical and clinical studies have also evidenced that these compounds show potent anti-colon cancer effects and exhibit negligible toxicity towards the normal cells. Hence, the present evidence-based study aimed to provide an update on various alkaloids that have been reported to induce ROS-mediated apoptosis in colon cancer cells via targeting various cellular components including hormones and growth factors, which play a role in metastasis, angiogenesis, proliferation, and invasion. This study also provides an individual account on each such alkaloid that underwent clinical trials either alone or in combination with other clinical drugs. In addition, various classes of phytochemicals that induce ROS-mediated cell death in different kinds of cancers including colon cancer are discussed.
Collapse
Affiliation(s)
- Vinod K. Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Juturu Mastanaiah
- Department of Pharmacology, Balaji College of Pharmacy, Anantapur, India
| | | | - Geetha Birudala
- Faculty of Pharmacy, Dr. M.G.R. Educational and Research Institute, Chennai, India
| | - Yahya F. Jamous
- Vaccines and Bioprocessing Centre, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Omar Alshargi
- College of Pharmacy, Riyadh ELM University, Riyadh, Saudi Arabia
| | - Kranthi Kumar Kotha
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bengaluru, India
| | - Hari Hara Sudhan
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | | | | | | | | | | | | | - Vinyas Mayasa
- GITAM School of Pharmacy, GITAM University Hyderabad Campus, Rudraram, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
| | - Adriana Kolesarova
- Faculty of Biotechnology and Food Sciences, Slovak University of Agriculture in Nitra, Nitra, Slovakia
| | - Petr Slama
- Laboratory of Animal Immunology and Biotechnology, Department of Animal Morphology, Physiology and Genetics, Faculty of AgriSciences, Mendel University in Brno, Brno, Czechia
| | | |
Collapse
|
50
|
Ukkola I, Nummela P, Heiskanen A, Holm M, Zafar S, Kero M, Haglund C, Satomaa T, Kytölä S, Ristimäki A. N-Glycomic Profiling of Microsatellite Unstable Colorectal Cancer. Cancers (Basel) 2023; 15:3571. [PMID: 37509233 PMCID: PMC10376987 DOI: 10.3390/cancers15143571] [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: 06/12/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Aberrant glycosylation affects cancer progression and immune evasion. Approximately 15% of colorectal cancers (CRCs) demonstrate microsatellite instability (MSI) and display major differences in outcomes and therapeutic responses, as compared to corresponding microsatellite stable (MSS) tumors. We compared the N-glycan profiles of stage II and IV MSI CRC tumors, further subdivided into BRAFV600E wild-type and mutated subgroups (n = 10 in each subgroup), with each other and with those of paired non-neoplastic mucosal samples using mass spectrometry. Further, the N-glycans of BRAFV600E wild-type stage II MSI tumors were compared to corresponding MSS tumors (n = 9). Multiple differences in N-glycan profiles were identified between the MSI CRCs and control tissues, as well as between the stage II MSI and MSS samples. The MSI CRC tumors showed a lower relative abundance of high-mannose N-glycans than did the control tissues or the MSS CRCs. Among MSI CRC subgroups, acidic N-glycans showed tumor stage and BRAF mutation status-dependent variation. Specifically, the large, sulfated/phosphorylated, and putative terminal N-acetylhexosamine-containing acidic N-glycans differed between the MSI CRC subgroups, showing opposite changes in stages II and IV, when comparing BRAF mutated and wild-type tumors. Our results show that molecular subgroups of CRC exhibit characteristic glycan profiles that may explain certain carcinogenic properties of MSI tumors.
Collapse
Affiliation(s)
- Iiris Ukkola
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | - Pirjo Nummela
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | | | - Matilda Holm
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Department of Surgery, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Sadia Zafar
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | - Mia Kero
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Caj Haglund
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Department of Surgery, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Tero Satomaa
- Glykos Finland Co., Ltd., 00790 Helsinki, Finland
| | - Soili Kytölä
- HUSLAB, Department of Genetics, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Ari Ristimäki
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|