Journal of Cancer Immunology & Therapy

All submissions of the EM system will be redirected to Online Manuscript Submission System. Authors are requested to submit articles directly to Online Manuscript Submission System of respective journal.
Reach Us +1 (202) 780-3397

Perspective - Journal of Cancer Immunology & Therapy (2023) Volume 6, Issue 3

The role of tumor microenvironment in shaping anti-tumor immune responses.

John Maxwell*

Department of Medical Oncology, National Cancer Centre, Singapore

*Corresponding Author:
John Maxwell
Department of Medical Oncology
National Cancer Centre
Singapore
E-mail:john.max52@gmail.com

Received:17-May-2023,Manuscript No. AAJCIT-23-102145; Editor assigned: 19-May-2023, PreQC No. AAJCIT-23-102145(PQ); Reviewed:03-Jun-2023, QC No. AAJCIT-23-102145; Revised:07-Jun-2023, Manuscript No. AAJCIT-23-102145(R); Published:14-Jun-2023, DOI: 10.35841/aajcit- 6.3.148

Citation: Maxwell J. The role of tumor microenvironment in shaping anti-tumor immune responses. J Cancer Immunol Ther. 2023;6(3):148

Visit for more related articles at Journal of Cancer Immunology & Therapy

Abstract

  

Introduction

The interaction between tumours and the immune system is a complex and dynamic process that greatly influences the development and progression of cancer. The tumor microenvironment (TME) is a unique ecosystem surrounding the tumor, composed of various cellular and non-cellular components. In recent years, there has been a growing understanding of the critical role played by the TME in shaping anti-tumor immune responses. This article explores the diverse components of the TME and highlights their impact on the immune system's ability to recognize and eliminate cancer cells .

Cellular components of the tumor microenvironment

The TME consists of several cellular components, including immune cells, fibroblasts, endothelial cells, and tumor-associated macrophages (TAMs). These cells actively interact and communicate with each other, impacting the immune response against tumours [1].

Immune cells: Immune cells such as T cells, Natural Killer (NK) Cells, Dendritic Cells (DCs), and Myeloid-Derived Suppressor Cells (MDSCs) play a crucial role in anti-tumor immunity. However, the TME can modulate their function and phenotype. For instance, T regulatory (Treg) cells, a subset of T cells, can suppress the immune response and promote tumor growth. Tumor-Infiltrating Lymphocytes (TILs) can also exhibit an exhausted phenotype, compromising their anti-tumor activity.

TAMs: TAMs, a type of immune cell, are often skewed towards a pro-tumoral phenotype in the TME. They can secrete immunosuppressive molecules, promote angiogenesis, and create an immunosuppressive microenvironment. Reprogramming TAMs towards an anti-tumor phenotype could enhance immune responses against tumors.

Fibroblasts: Cancer-Associated Fibroblasts (CAFs) are key components of the TME. They contribute to tumor growth and metastasis by producing Extracellular Matrix (ECM) components and growth factors. CAFs can also modulate the immune response by promoting immunosuppression and inhibiting T cell infiltration[2].

Non-cellular components of the tumor microenvironment

The non-cellular components of the TME include the ECM, cytokines, chemokines, and metabolites. These components actively participate in shaping anti-tumor immune responses.

ECM: The ECM provides structural support to the tumor and influences various processes, including cell migration, invasion, and angiogenesis. It can also act as a physical barrier, hindering immune cell infiltration into the tumor. Remodelling the ECM to facilitate immune cell infiltration has emerged as a potential therapeutic strategy.

Cytokines and chemokines: The TME contains a plethora of cytokines and chemokines that regulate immune cell recruitment, activation, and differentiation. Tumor-derived factors such as TGF-β and IL-10 can suppress immune responses, while pro-inflammatory cytokines like IFN-γ can enhance anti-tumor immunity. Understanding the balance and interactions of these molecules is crucial for effective immunotherapy [3].

Metabolites: Metabolic alterations in the TME can impact immune cell function. Tumor cells often exhibit increased glucose consumption and lactate production, leading to an acidic TME. This acidic environment can impair T cell function and promote immune suppression. Additionally, metabolic waste products, such as adenosine, can inhibit immune responses.

Therapeutic implications and future directions

Recognizing the significant influence of the TME on anti-tumor immune responses has led to the development of novel therapeutic strategies targeting the TME.

Immunotherapies: Immune checkpoint inhibitors (ICIs), such as anti-PD-1 and anti-CTLA-4 antibodies, have shown remarkable success in treating certain cancers by reinvigorating exhausted T cells. Combination therapies, including ICIs and agents targeting components of the TME, aim to enhance the effectiveness of immunotherapy.

Stromal targeting: Targeting stromal cells, such as CAFs, and ECM components holds promise in modulating the TME to support anti-tumor immune responses. Preclinical studies targeting CAFs or specific ECM components have shown encouraging results, warranting further investigation [4].

Metabolic interventions: Strategies aimed at modulating the metabolic profile of the TME have gained attention. These include inhibiting metabolic checkpoints and reprogramming tumor metabolism to enhance immune cell function. Clinical trials exploring these approaches are on-going [5].

Conclusion

The tumor microenvironment plays a crucial role in shaping anti-tumor immune responses. The complex interplay between the cellular and non-cellular components of the TME can either promote or suppress immune surveillance and the eradication of cancer cells. Understanding the mechanisms by which the TME influences immune responses is crucial for developing effective cancer immunotherapies. Targeting the TME, either independently or in combination with immunotherapies, holds significant promise for improving patient outcomes. Further research and clinical investigations are necessary to unravel the complexities of the TME and harness its potential for developing innovative cancer treatments.

References

  1. Euvrard S, Kanitakis J, Claudy A, et al.Skin cancers after organ transplantation. N Eng J Med. 2003;348(17):1681-91.
  2. Indexed at, Google Scholar, Cross Ref

  3. Stoff B, Salisbury C, Parker D, et al. Dermatopathology of skin cancer in solid organ transplant recipients. Transp Rev. 2010;24(4):172-89.
  4. Indexed at, Google Scholar, Cross Ref

  5. Van den Broek ME, Kägi D, Ossendorp F, et al. Decreased tumor surveillance in perforin-deficient mice. J Exp Med. 1996;184(5):1781-90.
  6. Indexed at, Google Scholar, Cross Ref

  7. Engel AM, Svane IM, Rygaard J, et al.MCA sarcomas induced in scid mice are more immunogenic than MCA sarcomas induced in congenic, immunocompetent mice. Scand J Immunol. 1997;45(4):463-70.
  8. Indexed at, Google Scholar, Cross Ref

  9. Aggarwal BB, Shishodia S, Sandur SK, et al.Inflammation and cancer: How hot is the link?. Biochem Pharmacol. 2006;72(11):1605-21.
  10. Indexed at, Google Scholar,Cross Ref

Get the App