null

CD39: A Novel Target to Overcome Immunosuppression in Cancer

Introduction to CD39 and Cancer Immunosuppression 


CD39 is an ectonucleotidase enzyme that plays a crucial role in generating adenosine, a powerful immunosuppressive molecule within the tumor microenvironment. By converting extracellular ATP (a danger signal that activates the immune system) into AMP, CD39 initiates the adenosine production pathway, which is completed by CD73. Adenosine, in turn, suppresses immune cell activity, particularly that of T cells, natural killer (NK) cells, and dendritic cells, thus protecting tumor cells from immune destruction.


Targeting CD39 with therapies like BU69, a monoclonal antibody that inhibits CD39 activity, is a promising strategy to reverse tumor-induced immunosuppression. By blocking CD39, these therapies prevent the formation of immunosuppressive adenosine, restoring immune function and enabling more robust anti-tumor responses. This article explores the biology of CD39, its role in the adenosine pathway, and the therapeutic potential of CD39-targeting immunotherapy.


CD39: Structure, Function, and the Adenosine Pathway 


The Role of CD39 in Adenosine Production


CD39 is a surface enzyme expressed on regulatory T cells (Tregs), macrophages, tumor cells, and other immune cells within the tumor microenvironment. It functions as the first step in the adenosine production pathway by hydrolyzing extracellular ATP into AMP. CD73, another ectonucleotidase, then converts AMP into adenosine, which has potent immunosuppressive effects when it binds to adenosine receptors (particularly A2A and A2B receptors) on immune cells.


This adenosine signaling inhibits the cytotoxic activity of effector immune cells and promotes the survival of tumor cells by dampening anti-tumor immune responses. CD39 and CD73 are often co-expressed in the tumor microenvironment, driving high adenosine levels that shield the tumor from immune attack.


CD39's Contribution to Tumor Immune Evasion


High levels of CD39 are commonly found in various cancers, including:

Tumor cells and immune cells, such as Tregs and myeloid-derived suppressor cells (MDSCs), use CD39 to generate immunosuppressive adenosine, effectively creating a barrier against immune activation. This process enables tumors to evade immune surveillance by:


  • Inhibiting T cell proliferation and function: Adenosine signaling via the A2A receptorsuppresses cytotoxic T lymphocytes (CTLs), which are responsible for killing tumor cells.
  • Reducing NK cell activity: Adenosine reduces the cytotoxic potential of NK cells, allowing tumors to grow unchecked.
  • Promoting Treg function: CD39-expressing Tregs suppress effector T cells and other immune responses, further contributing to tumor immune evasion.

Given its critical role in facilitating immunosuppression, CD39 has emerged as a key therapeutic target in cancer immunotherapy, with the goal of restoring immune cell function by inhibiting adenosine production.


Targeting CD39: A Strategy to Reverse Immunosuppression 


BU69: A Monoclonal Antibody Targeting CD39


BU69 is a monoclonal antibody that selectively binds to CD39, blocking its enzymatic activity and preventing the conversion of ATP into AMP. By inhibiting CD39, BU69 disrupts the adenosine production pathway, reducing the levels of immunosuppressive adenosine in the tumor microenvironment. This allows immune cells, including T cells and NK cells, to regain their anti-tumor activity.


The mechanism of action of BU69 can be summarized as follows:


  1. Inhibition of CD39 activity: BU69 binds to CD39, blocking its ability to hydrolyze ATP into AMP.
  2. Reduction of adenosine levels: With CD39 inhibited, less AMP is available for CD73 to convert into adenosine, resulting in lower adenosine concentrations in the tumor microenvironment.
  3. Restoration of immune function: The reduction in adenosine levels enables effector T cells and NK cells to recover their cytotoxic functions and attack tumor cells.
Synergy with other immunotherapies: BU69 can enhance the effectiveness of checkpoint inhibitors like anti-PD-1 and anti-CTLA-4 by removing adenosine-mediated suppression, allowing these therapies to fully activate immune responses.


Clinical Potential of BU69 in Cancer Therapy 


Targeting CD39 with BU69 is particularly promising in cancers where high adenosine levels contribute to immune resistance. Several cancers that express high levels of CD39 are candidates for BU69 therapy, including:


  • Breast cancer: CD39 expression on tumor cells and Tregs creates a highly immunosuppressive environment in breast cancer, making it resistant to immune attack.
  • Colorectal cancer: CD39 blockade could enhance immune infiltration and tumor destruction in colorectal cancer, where adenosine-mediated suppression is a key resistance mechanism.
  • Pancreatic cancer: Known for its highly immunosuppressive microenvironment, pancreatic cancer could benefit from CD39 targeting to reverse Treg- and adenosine-mediated suppression.
  • Ovarian cancer: In ovarian cancer, CD39 is expressed by both tumor cells and immune cells,
    promoting immune evasion. BU69 has the potential to reduce adenosine levels and enhance immune responses.

The therapeutic potential of BU69 extends to both solid tumors and hematologic malignancies where CD39 plays a role in immune evasion and tumor progression.


Synergistic Potential of CD39 Blockade in Combination Immunotherapy 


Enhancing the Efficacy of Checkpoint Inhibitors


One of the most exciting aspects of targeting CD39 is its potential to work synergistically with existing checkpoint inhibitors. Anti-PD-1 and anti-CTLA-4 therapies have shown remarkable success in treating various cancers, but their effectiveness is often limited by the presence of adenosine in the tumor microenvironment. By reducing adenosine production, CD39 inhibitors like BU69 can help to overcome this limitation.


  • Restoring T cell activity: Checkpoint inhibitors block immune checkpoints that inhibit T cell activation. However, in a high-adenosine environment, even activated T cells may be suppressed. BU69 can lower adenosine levels, allowing checkpoint inhibitors to fully activate T cells and sustain anti-tumor responses.
  • Overcoming immunotherapy resistance: Tumors that have developed resistance to checkpoint inhibitors often do so by upregulating immunosuppressive pathways like the adenosine pathway. Combining CD39 inhibition with checkpoint blockade offers a strategy to overcome this resistance and improve treatment outcomes.

CD39 Blockade and Chemotherapy


CD39 inhibition is also being explored in combination with chemotherapy. Chemotherapy often causes the release of ATP from dying tumor cells, which is then converted into AMP and adenosine by CD39 and CD73. Blocking CD39 can prevent this adenosine production, thereby enhancing the immune-stimulatory effects of chemotherapy and promoting a stronger anti-tumor immune response.


CD39 in the Tumor Microenvironment: A Dual Role 


While CD39's role in generating adenosine is well-established, it is also involved in other aspects of tumor biology. CD39 is expressed not only on tumor cells but also on various immune cells, including Tregs, macrophages, and myeloid-derived suppressor cells (MDSCs). These immune cells further contribute to the immunosuppressive milieu in the tumor microenvironment.


Tregs and CD39


Tregs, which are essential for maintaining immune tolerance, express high levels of CD39. In cancer, CD39-expressing Tregs inhibit effector T cells and promote tumor growth by suppressing anti-tumor immune responses. By targeting CD39 on Tregs, therapies like BU69 can reduce their suppressive activity, allowing cytotoxic T cells to attack the tumor more effectively.


Myeloid Cells and CD39


CD39 is also expressed on macrophages and MDSCs, which are known to play immunosuppressive roles in the tumor microenvironment. Blocking CD39 on these cells can reduce their ability to suppress immune responses, further contributing to the restoration of anti-tumor immunity. 


Challenges and Future Directions in CD39-Targeted Therapy 


Managing Immune-Related Toxicities


As with other immune-modulating therapies, targeting CD39 with BU69 carries the potential for immune-related adverse events (irAEs). These may include inflammatory responses and autoimmune-like reactions, as the immune system is reactivated to attack not only tumors but also healthy tissues. Future research will focus on optimizing dosing and identifying biomarkers to predict which patients are most likely to benefit from CD39 inhibition while minimizing irAEs. 


Overcoming Tumor Resistance 


While CD39 blockade holds significant promise, tumors may develop resistance by upregulating alternative immunosuppressive pathways. For example, tumor cells could increase the expression of CD73 or other ectonucleotidases to maintain adenosine production despite CD39 inhibition. Combination therapies that target both CD39 and CD73, or other immunosuppressive molecules, may be necessary to achieve sustained anti-tumor responses.


Conclusion 


CD39 is a key player in the adenosine-mediated immunosuppressive network that protects tumors from immune attack. Targeting CD39 with therapies like BU69 offers a promising approach to reverse immunosuppression, restore immune function, and enhance the efficacy of existing cancer immunotherapies. By blocking the production of adenosine, CD39 inhibitors have the potential to overcome resistance to checkpoint inhibitors and other treatments, providing a new avenue for combating refractory cancers. As research into CD39-targeted therapies progresses, it is poised to become a critical component of next-generation cancer immunotherapy strategies. 


References 


  1. Allard, B., et al., 2016. Targeting CD39 in cancer: Promising therapeutic approaches. Immunological Reviews, 276(1), pp.48-64.

  2. Stagg, J., et al., 2010. CD39 and CD73: Immunosuppressive ectonucleotidases in cancer. Cellular and Molecular Life Sciences, 67(4), pp.569-579.

  3. Bastid, J., et al., 2015. Targeting CD39 and CD73: A novel approach to cancer immunotherapy. Cancer Immunology, Immunotherapy, 64(11), pp.1373-1383.

  4. Antonioli, L., et al., 2013. Regulation of immunity by adenosine and adenosine deaminase in cancer. Oncogene, 32(14), pp.1873-1881.

  5. Hatfield, S.M., et al., 2015. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Science Translational Medicine, 7(277), pp.277-229.

  6. Perrot, I., et al., 2019. Blocking the adenosine A2A receptor potentiates CD73 blockade and immune checkpoint inhibition. Cancer Immunology Research, 7(3), pp.353-367.

  7. Yu, M., et al., 2019. Targeting CD39 to enhance cancer immunotherapy. Cancer Research, 79(1), pp.8-16.

  8. Vijayan, D., et al., 2017. Targeting the CD39-CD73 adenosine axis in cancer immunotherapy. Immunological Reviews, 276(1), pp.99-112.

21st Oct 2024 Zainab Riaz

Recent Posts