Tumor microenvironment signaling and therapeutics in cancer progression

Abstract Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion....

Full description

Bibliographic Details
Main Authors: Anshika Goenka, Fatima Khan, Bhupender Verma, Priyanka Sinha, Crismita C. Dmello, Manasi P. Jogalekar, Prakash Gangadaran, Byeong‐Cheol Ahn
Format: Article
Language:English
Published: Wiley 2023-05-01
Series:Cancer Communications
Subjects:
Online Access:https://doi.org/10.1002/cac2.12416
_version_ 1797829303236296704
author Anshika Goenka
Fatima Khan
Bhupender Verma
Priyanka Sinha
Crismita C. Dmello
Manasi P. Jogalekar
Prakash Gangadaran
Byeong‐Cheol Ahn
author_facet Anshika Goenka
Fatima Khan
Bhupender Verma
Priyanka Sinha
Crismita C. Dmello
Manasi P. Jogalekar
Prakash Gangadaran
Byeong‐Cheol Ahn
author_sort Anshika Goenka
collection DOAJ
description Abstract Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion. A deep understanding of the signaling pathways involved in cell‐to‐cell and cell‐to‐ECM interactions is needed to design effective intervention strategies that might interrupt these interactions. In this review, we describe the tumor microenvironment (TME) components and associated therapeutics. We discuss the clinical advances in the prevalent and newly discovered signaling pathways in the TME, the immune checkpoints and immunosuppressive chemokines, and currently used inhibitors targeting these pathways. These include both intrinsic and non‐autonomous tumor cell signaling pathways in the TME: protein kinase C (PKC) signaling, Notch, and transforming growth factor (TGF‐β) signaling, Endoplasmic Reticulum (ER) stress response, lactate signaling, Metabolic reprogramming, cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING) and Siglec signaling pathways. We also discuss the recent advances in Programmed Cell Death Protein 1 (PD‐1), Cytotoxic T‐Lymphocyte Associated Protein 4 (CTLA4), T‐cell immunoglobulin mucin‐3 (TIM‐3) and Lymphocyte Activating Gene 3 (LAG3) immune checkpoint inhibitors along with the C‐C chemokine receptor 4 (CCR4)‐ C‐C class chemokines 22 (CCL22)/ and 17 (CCL17), C‐C chemokine receptor type 2 (CCR2)‐ chemokine (C‐C motif) ligand 2 (CCL2), C‐C chemokine receptor type 5 (CCR5)‐ chemokine (C‐C motif) ligand 3 (CCL3) chemokine signaling axis in the TME. In addition, this review provides a holistic understanding of the TME as we discuss the three‐dimensional and microfluidic models of the TME, which are believed to recapitulate the original characteristics of the patient tumor and hence may be used as a platform to study new mechanisms and screen for various anti‐cancer therapies. We further discuss the systemic influences of gut microbiota in TME reprogramming and treatment response. Overall, this review provides a comprehensive analysis of the diverse and most critical signaling pathways in the TME, highlighting the associated newest and critical preclinical and clinical studies along with their underlying biology. We highlight the importance of the most recent technologies of microfluidics and lab‐on‐chip models for TME research and also present an overview of extrinsic factors, such as the inhabitant human microbiome, which have the potential to modulate TME biology and drug responses.
first_indexed 2024-04-09T13:18:19Z
format Article
id doaj.art-d8791161e3824e2a8f6f16b55281fab8
institution Directory Open Access Journal
issn 2523-3548
language English
last_indexed 2024-04-09T13:18:19Z
publishDate 2023-05-01
publisher Wiley
record_format Article
series Cancer Communications
spelling doaj.art-d8791161e3824e2a8f6f16b55281fab82023-05-11T15:21:49ZengWileyCancer Communications2523-35482023-05-0143552556110.1002/cac2.12416Tumor microenvironment signaling and therapeutics in cancer progressionAnshika Goenka0Fatima Khan1Bhupender Verma2Priyanka Sinha3Crismita C. Dmello4Manasi P. Jogalekar5Prakash Gangadaran6Byeong‐Cheol Ahn7The Ken & Ruth Davee Department of Neurology The Robert H. Lurie Comprehensive Cancer Center Northwestern University Feinberg School of Medicine Chicago, 60611 IL USADepartment of Neurological Surgery Feinberg School of Medicine Northwestern University Chicago, 60611 IL USADepartment of Ophthalmology Schepens Eye Research Institute Massachusetts Eye and Ear Infirmary Harvard Medical School Boston, 02114 MA USADepartment of Neurology MassGeneral Institute for Neurodegenerative Disease Massachusetts General Hospital, Harvard Medical School Charlestown, 02129 MA USADepartment of Neurological Surgery Feinberg School of Medicine Northwestern University Chicago, 60611 IL USAHelen Diller Family Comprehensive Cancer Center University of California San Francisco San Francisco, 94143 CA USABK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents Department of Biomedical Science, School of Medicine Kyungpook National University Daegu, 41944 South KoreaBK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents Department of Biomedical Science, School of Medicine Kyungpook National University Daegu, 41944 South KoreaAbstract Tumor development and metastasis are facilitated by the complex interactions between cancer cells and their microenvironment, which comprises stromal cells and extracellular matrix (ECM) components, among other factors. Stromal cells can adopt new phenotypes to promote tumor cell invasion. A deep understanding of the signaling pathways involved in cell‐to‐cell and cell‐to‐ECM interactions is needed to design effective intervention strategies that might interrupt these interactions. In this review, we describe the tumor microenvironment (TME) components and associated therapeutics. We discuss the clinical advances in the prevalent and newly discovered signaling pathways in the TME, the immune checkpoints and immunosuppressive chemokines, and currently used inhibitors targeting these pathways. These include both intrinsic and non‐autonomous tumor cell signaling pathways in the TME: protein kinase C (PKC) signaling, Notch, and transforming growth factor (TGF‐β) signaling, Endoplasmic Reticulum (ER) stress response, lactate signaling, Metabolic reprogramming, cyclic GMP–AMP synthase (cGAS)–stimulator of interferon genes (STING) and Siglec signaling pathways. We also discuss the recent advances in Programmed Cell Death Protein 1 (PD‐1), Cytotoxic T‐Lymphocyte Associated Protein 4 (CTLA4), T‐cell immunoglobulin mucin‐3 (TIM‐3) and Lymphocyte Activating Gene 3 (LAG3) immune checkpoint inhibitors along with the C‐C chemokine receptor 4 (CCR4)‐ C‐C class chemokines 22 (CCL22)/ and 17 (CCL17), C‐C chemokine receptor type 2 (CCR2)‐ chemokine (C‐C motif) ligand 2 (CCL2), C‐C chemokine receptor type 5 (CCR5)‐ chemokine (C‐C motif) ligand 3 (CCL3) chemokine signaling axis in the TME. In addition, this review provides a holistic understanding of the TME as we discuss the three‐dimensional and microfluidic models of the TME, which are believed to recapitulate the original characteristics of the patient tumor and hence may be used as a platform to study new mechanisms and screen for various anti‐cancer therapies. We further discuss the systemic influences of gut microbiota in TME reprogramming and treatment response. Overall, this review provides a comprehensive analysis of the diverse and most critical signaling pathways in the TME, highlighting the associated newest and critical preclinical and clinical studies along with their underlying biology. We highlight the importance of the most recent technologies of microfluidics and lab‐on‐chip models for TME research and also present an overview of extrinsic factors, such as the inhabitant human microbiome, which have the potential to modulate TME biology and drug responses.https://doi.org/10.1002/cac2.124163D‐modelcancer therapygut microbiotaimmune signalingmetabolismsignaling
spellingShingle Anshika Goenka
Fatima Khan
Bhupender Verma
Priyanka Sinha
Crismita C. Dmello
Manasi P. Jogalekar
Prakash Gangadaran
Byeong‐Cheol Ahn
Tumor microenvironment signaling and therapeutics in cancer progression
Cancer Communications
3D‐model
cancer therapy
gut microbiota
immune signaling
metabolism
signaling
title Tumor microenvironment signaling and therapeutics in cancer progression
title_full Tumor microenvironment signaling and therapeutics in cancer progression
title_fullStr Tumor microenvironment signaling and therapeutics in cancer progression
title_full_unstemmed Tumor microenvironment signaling and therapeutics in cancer progression
title_short Tumor microenvironment signaling and therapeutics in cancer progression
title_sort tumor microenvironment signaling and therapeutics in cancer progression
topic 3D‐model
cancer therapy
gut microbiota
immune signaling
metabolism
signaling
url https://doi.org/10.1002/cac2.12416
work_keys_str_mv AT anshikagoenka tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT fatimakhan tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT bhupenderverma tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT priyankasinha tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT crismitacdmello tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT manasipjogalekar tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT prakashgangadaran tumormicroenvironmentsignalingandtherapeuticsincancerprogression
AT byeongcheolahn tumormicroenvironmentsignalingandtherapeuticsincancerprogression