Checkpoint Inhibitor Therapy
Checkpoint Inhibitor Therapy
Checkpoint inhibitors are a revolutionary class of drugs that 'release the brakes' on the immune system, allowing it to mount a strong attack against cancer. Normally, the body uses certain proteins (like PD-1, PD-L1, and CTLA-4) as 'checkpoints' to prevent the immune system from becoming too aggressive and attacking healthy cells. However, cancer cells exploit these checkpoints to evade detection.
Checkpoint inhibitors, such as PD-L1 inhibitors (pembrolizumab, nivolumab) and CTLA-4 inhibitors (ipilimumab), work by blocking these proteins, freeing the immune system to recognise and attack cancer cells. By disrupting these checkpoints, checkpoint inhibitors unleash the immune system's full potential, often leading to lasting cancer remission, especially when combined with other therapies.
How Checkpoint Inhibitor Therapy Works
PD-L1/PD-1 Inhibition
PD-L1 is expressed in tumour cells and binds to PD-1 receptors on immune cells, effectively camouflaging the tumour. PD-L1 inhibitors block this interaction, allowing the immune cells to recognise and destroy the cancer.
CTLA-4 Blockade
CTLA-4, another immune checkpoint, regulates the immune system's response to ensure it doesn't overreact. Blocking CTLA-4 (with drugs like ipilimumab) enhances the immune response, making it more aggressive against cancer.
Understanding how checkpoint inhibitors work involves appreciating how the immune system targets and destroys cancer cells. To simplify this, we use the analogy of soldiers (immune cells) being equipped for battle against an enemy (cancer cells). The following are the five critical aspects of this process:
Number of Immune Cells
Increasing the number of immune cells, particularly natural killer cells, is crucial for mounting a strong immune response. Therapies like GC-MSF, mushroom extracts, and herbal supplements such as astragalus and reishi help boost these cell numbers.
Activity of Immune Cells
It’s not just about having enough immune cells; their activity must also be heightened. Treatments like photodynamic therapy, electro-hyperthermia, and intravenous vitamin C can enhance immune cell activity, turning passive cells into active cancer fighters.
Tumour Penetration
The tumour microenvironment is often hostile, with barriers that prevent immune cells from reaching the cancer. By targeting this environment with treatments like angiogenesis inhibitors and hyperthermia, we can improve immune cell penetration, allowing them to reach and destroy cancer cells more effectively.
Cancer CEll Identification
Cancer cells evade detection by the immune system. Checkpoint inhibitors expose these hidden cells by blocking their PD-L1 and CTLA-4 proteins. Once identified, the immune system can attack and destroy them.
Tumour Cell Death
Once attacked, cancer cells must be prevented from repairing themselves. Treatments like PARP inhibitors stop cancer cells from recovering, ensuring their destruction through apoptosis.
The Advantages and Disadvantages of Checkpoint Inhibitor Therapy
One of the key benefits of checkpoint inhibitor therapy is its high effectiveness, particularly in cancers that have proven resistant to traditional therapies, with the potential to induce long-term remission in some patients.
This therapy also demonstrates strong compatibility with other treatments, including chemotherapy, radiotherapy, and other forms of immunotherapy, making it a versatile option within cancer treatment regimens. Additionally, compared to traditional approaches like chemotherapy, checkpoint inhibitors tend to cause milder and less frequent side effects, which can improve the overall quality of life for patients undergoing treatment.
Despite its benefits, checkpoint inhibitor therapy has certain drawbacks. The cost of long-term treatment can be substantial, which may pose challenges for accessibility and affordability. Moreover, although side effects are generally milder, there is a risk of rare but serious autoimmune reactions.
In such cases, the immune system may mistakenly target healthy organs, such as the thyroid, liver, or lungs, leading to potentially severe complications. These factors underscore the need for careful patient monitoring and consideration when utilising this form of immunotherapy.
Our Approach to Checkpoint Inhibitor Therapy
At Sanctura, we thoroughly assess the characteristics of each patient’s tumour, looking for factors such as tumour mutational burden, microsatellite instability, and the expression of PD-L1 and PD-1, which determine how likely immunotherapy is to work. This personalised approach ensures that our patients receive the most effective treatment tailored to their cancer's specific needs.
Targeted Monoclonal Antibodies
Targeted Monoclonal Antibodies
Targeted monoclonal antibodies (mAbs) are a cornerstone of cancer immunotherapy, offering tailored treatment options that leverage the body’s immune system to fight cancer more effectively while sparing healthy tissue. They are laboratory-produced molecules engineered to mimic the immune system's ability to fight cancer by targeting antigens on cancer cells or the surrounding environment.
Here's how they work in the context of immunotherapy:
How Targeted Monoclonal Antibodies Work
Target Specific Antigens
Monoclonal antibodies are designed to bind to specific proteins (antigens) that are overexpressed on the surface of cancer cells or other cells involved in tumour growth. For example, HER2 is a protein overexpressed in some breast cancers, and trastuzumab (Herceptin) is a monoclonal antibody specifically targeting HER2.
Mechanisms of Action
Some mAbs flag cancer cells for destruction by immune cells, such as natural killer (NK) cells or macrophages, through a process called antibody-dependent cellular cytotoxicity (ADCC).
Certain mAbs inhibit growth signals that cancer cells need to proliferate. For example, cetuximab targets the epidermal growth factor receptor (EGFR) to stop signalling pathways that promote tumour growth.
Conjugated mAbs are linked to drugs, toxins, or radioactive substances delivered directly to cancer cells, sparing healthy cells. These are known as antibody-drug conjugates (ADCs).
Some mAbs, like immune checkpoint inhibitors (e.g., pembrolizumab), block proteins such as PD-1 or CTLA-4 that suppress immune responses, reactivating the immune system to attack tumours.
Some monoclonal antibodies are even bi-specific, meaning they are engineered to target two different antigens simultaneously. For instance, they may bring T-cells in direct contact with cancer cells to enhance immune killing.
Advantages of Targeted Monoclonal Antibodies
Precision
Minimises damage to normal cells, reducing side effects compared to traditional chemotherapy.
Versatility
Can be used alone or with other therapies like chemotherapy, radiation, or immune checkpoint inhibitors.
Immune Engagement
Enhances the natural immune response against cancer cells.
Challenges of Targeted Monoclonal Antibodies
Resistance
Cancer cells can mutate, altering the target antigen or bypassing the pathways affected by mAbs.
Toxicity
Some monoclonal antibodies can still cause immune-related side effects or off-target effects.
Cost
mAb treatments are expensive due to their development and production complexity.
Examples of Targeted Monoclonal Antibodies in Cancer Treatment
HER2-Positive Breast Cancer
Trastuzumab targets the HER2 receptor.
Non-Hodgkin Lymphoma
Rituximab binds to CD20 on B-cells.
Lung Cancer
Bevacizumab targets VEGF to inhibit angiogenesis (blood vessel formation).
Melanoma and Lung Cancer
Checkpoint inhibitors like nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4).