echemi logo
Product
  • Product
  • Supplier
  • Inquiry
    Home > Active Ingredient News > Antitumor Therapy > Systemic immune landscape of cancer

    Systemic immune landscape of cancer

    • Last Update: 2023-01-04
    • Source: Internet
    • Author: User
    Search more information of high quality chemicals, good prices and reliable suppliers, visit www.echemi.com

     

    preface

    Cancer is a systemic disease, and long-term inflammation is one of the main hallmarks of
    cancer.
    Whether this inflammation triggers tumorigenesis or supports tumor growth depends on the environment, but ultimately the systemic immune landscape beyond the tumor changes significantly during tumor progression
    .

    The field of tumor immunology focuses on local immune responses in the tumor microenvironment (TME), whereas immunity is coordinated across tissues
    .
    Without continuous communication with the periphery, a local anti-tumor immune response cannot be present
    .
    In addition, almost every subset of immune cells is involved
    in cancer biology.
    Therefore, a thorough understanding of the cancer immune response must include the entire peripheral immune system as well as all immune cell lineages
    within TME.

     

    Perturbations caused by tumor burden

    Many human and mouse cancer models lead to widespread disruption
    of hematopoietic function.
    This disruption is most pronounced by the expansion of immature neutrophils and monocytes in the periphery of the tumor host, which then enter TME and lead to local immunosuppression
    .

    Hematopoietic stem cells and progenitor cells are mobilized into the proliferation and differentiation of monocytes and granulocyte lines, resulting in peripheral expansion and intratumor accumulation of immature immunosuppressive neutrophils, including polymorphonuclear myelo-derived suppressor cells (PMN-MDSCs), monocytes (M-MDSCs), and macrophages
    。 A comprehensive meta-analysis of more than 40,000 patients found that elevated neutrophil frequency in the blood, as measured by neutrophil-to-lymphocyte ratio, was associated with poor outcomes in patients with mesothelioma, pancreatic, renal cell carcinoma, colorectal cancer, gastroesophageal cancer, non-small cell lung cancer (
    NSCLC), cholangiocarcinoma, and hepatocellular carcinoma.

    In addition to the overproduction of monocytes and neutrophils by abnormal hematopoiesis, dendritic cell perturbations
    are observed on the periphery of the tumor-burdening host.
    This has important implications for the development of anti-tumor immune responses, as dendritic cells are in many cases key coordinators
    for the initiation, differentiation and proliferation of CD8+ and CD4+ T cells.
    Cancer patients have fewer
    DC cells in their peripheral blood compared to healthy controls.

    The perturbation of T cells in the most studied cancer is the expansion of peripheral inhibitory CD4+ regulatory T (Treg) cells and their invasion into the tumor
    .
    Recent studies have shown that Treg cells in the blood of cancer patients have the same phenotype and TCR profile as intratumor T cells, suggesting that a significant proportion of intratumor suppressor Treg cells are derived from naturally occurring thymic Treg cells rather than differentiated by tumor-induced naïve CD4+ T cells
    .

    Another suppressor lymphocytes that play a role in tumor progression are regulatory B cells, which are characterized by the production of the anti-inflammatory cytokine IL-10
    .
    Similar to Treg cells, expansion of regulatory B cells is observed in the peripheral blood of patients with gastric and lung cancer, while the frequency of total B cells remains unchanged
    .

    In addition, natural killer (NK) cells are another important component of
    anti-tumor immunity.
    Peripheral NK cells in breast cancer patients also have an altered phenotype characterized by decreased expression of activating receptors, including NKp30, NKG2D, DNAM-1, and CD16, and in patients with gastrointestinal stromal tumors, peripheral NK cells show reduced expression levels of the activation receptor NKp30, and degranulation is impaired
    after NKp30 crosslinking.

     

    Changes in the immune system caused by traditional treatments

    Traditional treatment strategies for cancer, including chemotherapy, radiation and surgery, can also disrupt the immune landscape
    throughout the body.
    Understanding these systemic immune consequences is important
    for designing strategies that enhance, rather than hinder, the anti-tumor immune response.

    Chemotherapy and radiotherapy

    Chemotherapy and radiation therapy aim to target cancer cells
    by disrupting cell integrity during division.
    However, these drugs can also induce immune remodeling that hinders or enhances the overall therapeutic effect
    .

    The effects of chemotherapy and radiation on the immune system largely depend on the environment
    .
    In non-small cell lung cancer, standard, prolonged low-dose radiation therapy results in myeloid cell expansion, reduced antigen-presenting cell function, and impaired
    T cell response.
    Similar immune effects
    were observed in cervical cancer patients after combination chemotherapy and radiotherapy.

    Chemotherapy can enhance systemic anti-tumor immunity while disrupting cancer cell division
    .
    Recent studies have shown that
    an effective response to preoperative neoadjuvant chemotherapy for triple-negative breast cancer (TNBC) induces the recruitment of new T cell clones to TME rather than the expansion of original cells
    .
    In addition, different breast cancer subtypes exhibit different immune responses to chemotherapy, reflected in the
    function of peripheral CD8+ T cells.
    Patients with estrogen receptor-positive (
    ER+) breast tumors had decreased function of circulating PD1+CD8+ T cells, while patients with ER+HER2+ breast tumors had complete loss of function
    in this subgroup.
    In contrast, patients with TNBC exhibit elevated high-functioning PD1+CD8+ T cells, producing effector cytokines, including IFN-γ, TNF, and granzyme B, with evidence of
    clonal amplification.

    Tumor removal

    Several recent studies have shown that systemic wound healing induces myeloid immune cell remodeling
    .
    Resection of wounds that do not depend on the removal of the primary tumor can trigger healing, improve circulating IL-6, G-CSF, and CCL2, and ultimately push myeloid subsets into immunosuppressive states
    .

    However, there is evidence that primary tumors may be a major driver of systemic immune
    remodeling.
    Successful excision of the primary tumor in mouse models of breast and colon cancer is sufficient to restore normal systemic immune tissue to a large extent, making immune cells comparable
    to that of healthy control mice.

    Therefore, surgery can have harmful and beneficial effects on
    the systemic immune system.
    Immunosuppressive mechanisms triggered by early postoperative wound healing may provide a window
    of opportunity for cancer cell growth.
    However, a reduction in the burden of primary tumors can eventually restore systemic immunity, resulting in a strong adaptive response
    .
    It will be important
    to discover how cancer type, especially disease stage, affects immune reconstitution after surgery and the resulting metastatic potential.

     

    Systemic responses in immunotherapy

    The dominant view of the effectiveness of cancer immunotherapy revolves around the concept of revitalizing cytotoxic effectors within TME, but the fundamental systemic understanding of effective anti-tumor immunity is growing
    in this field.
    Recent studies have shown that immune checkpoint inhibitors (
    ICIs) rely on systemic immune mechanisms to achieve potent anti-tumor responses
    .
    In addition, the microbiome is becoming an effective regulator of the immune system
    .

    Complete peripheral immunity is essential for immunotherapy effectiveness

    Complete peripheral immune function, communication, and transport are necessary for
    ICI to be effective.
    Systemic chemotherapy may disrupt peripheral immune integrity, thereby hindering the therapeutic effect of PD-1 blockade, leading to systemic lymphatic depletion and eliminating long-term immune memory
    .
    In contrast, local chemotherapy avoids damaging peripheral immunity and, in synergy with PD-1 blockade, induces dendritic cell infiltration into tumors and clonal expansion of antigen-specific effector T cells
    .

    CD103+ dendritic cells migrate from tumors to dLN via a CCR7-dependent mechanism to transport tumor antigens to the peripheral immune system, and these dendritic cells can then also excite tumor-specific T cells
    .
    Newly started tumor-specific T cells then flow from lymph nodes to tumors, and this cycle is an important process
    in natural and therapeutically induced anti-tumor immunity.

    Effective immunotherapy drives a new immune response

    The antitumor response ultimately requires functional effector lymphocytes within TME to mediate cancer cell
    killing.
    However, over time, intratumor T cells may become depleted, preventing them from performing key effector functions
    .

    To overcome local immune dysfunction, effective immunotherapy drives a new peripheral immune response, ultimately leading to new effector T cell infiltration
    .
    Some reports have now shown that PD-1 and PD-L1 blockade can drive new T cell clones into TME that did not exist
    prior to treatment.
    In addition, anti-CTLA-4 has also been shown to significantly increase peripheral T cell responsiveness in melanoma patients, suggesting that novel T cell priming is a mechanism of
    action.

    Together, these results support the idea that not only is peripheral immunity involved in the novel antitumor response, but that de novo initiation of additional naïve T cells with neoantigen specificity also contributes to effective immunotherapy
    .

     

    brief summary

    In addition to the restructuring of the immune system in cancer, there is growing evidence that the immune state of tumor burden differs
    from the function of the immune system that is not disturbed.
    The development of a peripherally coordinated de novo anti-tumor immune response is critical
    for immunotherapy effectiveness.
    Any functional abnormalities in the tumor's immune system can lead to poor immunotherapy
    .
    Therefore, it is necessary for us to conduct more in-depth research on the global system immunity in cancer, which will help us develop tumor immune drugs and discover
    new targets.

    References:

    1.
    Systemic immunity in cancer.
    Nat Rev Cancer.
    2021; 21(6): 345–359.

     

    This article is an English version of an article which is originally in the Chinese language on echemi.com and is provided for information purposes only. This website makes no representation or warranty of any kind, either expressed or implied, as to the accuracy, completeness ownership or reliability of the article or any translations thereof. If you have any concerns or complaints relating to the article, please send an email, providing a detailed description of the concern or complaint, to service@echemi.com. A staff member will contact you within 5 working days. Once verified, infringing content will be removed immediately.

    Contact Us

    The source of this page with content of products and services is from Internet, which doesn't represent ECHEMI's opinion. If you have any queries, please write to service@echemi.com. It will be replied within 5 days.

    Moreover, if you find any instances of plagiarism from the page, please send email to service@echemi.com with relevant evidence.