GET THE APP

Unveiling the Complex Nexus: Inflammation′s Impact on Cancer Initiation, Progression and Therapy
..

Journal of Inflammatory Bowel Diseases & Disorders

ISSN: 2476-1958

Open Access

Perspective - (2023) Volume 8, Issue 3

Unveiling the Complex Nexus: Inflammation′s Impact on Cancer Initiation, Progression and Therapy

Floria Cheng*
*Correspondence: Floria Cheng, Department of Biomedical Technology Center, University of Muenster, Muenster, Germany, Email:
Department of Biomedical Technology Center, University of Muenster, Muenster, Germany

Received: 31-Jul-2023, Manuscript No. jibdd-23-111234; Editor assigned: 02-Aug-2023, Pre QC No. P-111234; Reviewed: 17-Aug-2023, QC No. Q-111234; Revised: 22-Aug-2023, Manuscript No. R-111234; Published: 29-Aug-2023 , DOI: 10.37421/2476-1958.2023.8.191
Citation: Cheng, Floria. “Unveiling the Complex Nexus: Inflammation's Impact on Cancer Initiation, Progression and Therapy.” J Inflamm Bowel Dis Disorder 8 (2023): 191.
Copyright: © 2023 Cheng F. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Introduction

In recent years, a growing body of research has highlighted the intricate relationship between inflammation and cancer. While inflammation is a natural immune response that aids in tissue repair and defense against pathogens, chronic or sustained inflammation can create an environment conducive to cancer initiation and progression. This article delves into the multifaceted connection between inflammation and cancer, exploring how inflammatory processes contribute to different stages of cancer development and the innovative therapeutic strategies being developed to disrupt this complex nexus. The recognition of inflammation's pivotal role in cancer has spurred the development of innovative therapeutic strategies that target the inflammatory microenvironment. Immune checkpoint inhibitors have emerged as game-changers by releasing the brakes on immune responses and enabling immune cells to recognize and attack cancer cells. Targeting key inflammatory pathways, such as NF-κB, holds promise for disrupting the intricate network of pro-inflammatory signals that promote cancer growth [1].

Description

Chronic inflammation can lead to DNA damage, mutations, and epigenetic changes that are key drivers of cancer initiation. Inflammatory cells release Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS), which can cause genetic alterations. Additionally, sustained inflammation can disrupt the normal tissue microenvironment, promoting the survival of cells with oncogenic mutations. During the promotion phase, inflammatory cytokines and chemokine’s attract immune cells to the site of inflammation. These immune cells release growth factors and signaling molecules that promote cell proliferation and angiogenesis. This creates an environment conducive to tumor growth and expansion. Inflammatory mediators facilitate tumor invasion, migration, and metastasis by promoting angiogenesis and remodeling of the extracellular matrix. Immune cells can also suppress the anti-tumor immune response, allowing cancer cells to evade immune surveillance [2,3].

The heterogeneity of both inflammation and cancer necessitates personalized approaches to treatment. Biomarkers that signify inflammatory activity within tumors can guide therapeutic decisions, allowing for tailored interventions that address the unique characteristics of each patient's disease. Combining anti-inflammatory drugs with other modalities, such as radiation and chemotherapy, holds the potential to enhance treatment efficacy by modifying the tumor microenvironment and sensitizing cancer cells to therapy.

NF-κB (Nuclear Factor-κB) is a central regulator of inflammation and is often deregulated in cancer cells. It promotes the expression of pro-inflammatory genes and contributes to tumor cell survival, growth, and invasion. Inflammatory cytokines such as IL-6, IL-1β and TNF-α play a pivotal role in fostering an inflammatory microenvironment within tumors. They contribute to cancer cell proliferation, angiogenesis, and metastasis. Macrophages, neutrophils and T cells infiltrate tumors in response to inflammation. However, tumor-associated immune cells can exhibit both pro-tumor and anti-tumor functions, creating a complex interplay in the tumor microenvironment. Nonsteroidal Anti-Inflammatory Drugs (NSAIDs) and corticosteroids have been investigated for their potential to inhibit inflammationassociated cancer. They may reduce the production of inflammatory mediators and disrupt the pro-tumor inflammatory cascade. Immune checkpoint inhibitors have shown remarkable success in enhancing anti-tumor immune responses. These therapies aim to unleash the immune system's ability to target cancer cells and overcome immunosuppressive signals in the tumor microenvironment. Inhibiting key signaling pathways involved in inflammation, such as NF-κB, holds promise for disrupting the link between chronic inflammation and cancer development [4,5].

Conclusion

The intricate interplay between inflammation and cancer reveals a complex nexus that spans cancer initiation, progression, and therapy response. As our understanding of the inflammatory-cancer connection deepens, novel therapeutic strategies are emerging to target inflammatory processes and reshape the tumor microenvironment. From anti-inflammatory agents to immunomodulatory therapies, these approaches offer hope for more effective cancer treatments and personalized interventions that harness the power of the immune system to combat cancer. Chronic inflammation, driven by cytokines, chemokines, and immune cells, plays a critical role in the initiation of genetic mutations and epigenetic changes that can lead to cancer. Moreover, the sustained release of inflammatory mediators creates an environment that supports tumor growth, angiogenesis, and metastasis. While inflammation can activate oncogenic pathways, it can also hinder anti-tumor immune responses, allowing cancer cells to evade surveillance.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Marceca, Gioacchino P., Priya Londhe and Federica Calore. "Management of cancer cachexia: Attempting to develop new pharmacological agents for new effective therapeutic options." Front Oncol 10 (2020): 298.
  2. Google Scholar, Crossref, Indexed at

  3. Lanser, Lukas, Patricia Kink, Eva Maria Egger and Wolfgang Willenbacher, et al. "Inflammation-induced tryptophan breakdown is related with anemia, fatigue and depression in cancer." Front Immunol 11 (2020): 249
  4.  Google Scholar, Crossref, Indexed at

  5. Laviano, Alessandro, Michael M. Meguid, Antonia Cascino and Alessio Molfino, et al. "Tryptophan in wasting diseases: At the crossing between immune function and behaviour." Curr Opin Clin Nutr Metab Care 12 (2009): 392-397.
  6. Google Scholar, Crossref, Indexed at

  7. Akagawa, Mitsugu. "Protein carbonylation: Molecular mechanisms, biological implications and analytical approaches." Free Radic Res 55 (2021): 307-320.
  8.  Google Scholar, Crossref, Indexed at

  9. Ábrigo, Johanna, Alvaro A. Elorza, Claudia A. Riedel and Cristian Vilos, et al. "Role of oxidative stress as key regulator of muscle wasting during cachexia." Oxid Med Cell Longev 2018 (2018).
  10. Google Scholar, Crossref, Indexed at

Google Scholar citation report
Citations: 77

Journal of Inflammatory Bowel Diseases & Disorders received 77 citations as per Google Scholar report

Journal of Inflammatory Bowel Diseases & Disorders peer review process verified at publons

Indexed In

 
arrow_upward arrow_upward