Skip to content

How Does Age Affect Immune Checkpoint Therapy Biomarkers?

5 min read

According to research, the majority of new cancer diagnoses occur in patients over the age of 65, making the study of age-related immune changes critical. These changes profoundly influence the biomarkers used to predict response to immune checkpoint therapy, affecting treatment strategies and outcomes.

Quick Summary

Age significantly impacts immune checkpoint therapy biomarkers by altering the immune landscape through immunosenescence, which can increase inhibitory molecule expression while potentially reducing T cell function and diversity. These complex changes create a different biomarker profile in older patients, necessitating a personalized approach to immunotherapy.

Key Points

  • Immunosenescence impacts biomarker reliability: The age-related remodeling of the immune system, known as immunosenescence, alters the expression and function of key biomarkers like PD-L1, complicating their predictive value in older patients.

  • Older patients have unique biomarker profiles: Older patients often show higher levels of inhibitory markers like PD-L1, LAG-3, and TIM-3, alongside a higher tumor mutational burden, which can create both challenges and opportunities for treatment.

  • Clinical outcomes are complex: While many studies show comparable immunotherapy efficacy across age groups, factors like patient frailty, comorbidities, and the specific biomarker profile can influence outcomes, sometimes suggesting reduced benefits for the very elderly.

  • Geriatric assessment is crucial: Chronological age alone is not a sufficient predictor of response. Comprehensive geriatric assessments are needed to determine an older patient's fitness for treatment and to personalize care.

  • Tumor microenvironment is influenced by age: Aging creates a more immunosuppressive tumor microenvironment, partly due to chronic inflammation and increased regulatory immune cells, which can dampen therapeutic response.

  • Future therapies may target age-specific pathways: Research is exploring novel strategies, such as combining immune checkpoint inhibitors with senolytic drugs to clear senescent cells, to overcome age-related immunotherapy resistance.

In This Article

The Foundation of Immune Checkpoint Therapy

Immune checkpoint therapy is a cornerstone of modern oncology, targeting specific proteins, or checkpoints, on immune and cancer cells to unleash the immune system's attack on tumors. In healthy individuals, these checkpoints maintain immune system balance, but cancer cells often hijack them to evade detection. The most studied biomarkers for these therapies include programmed death-ligand 1 (PD-L1), tumor mutational burden (TMB), and the status of mismatch repair deficiency (dMMR). However, the aging process introduces a new layer of complexity, reshaping the immune system in ways that can alter how these biomarkers behave and predict treatment efficacy.

Understanding Immunosenescence: The Aging Immune System

As the body ages, its immune system undergoes significant changes collectively known as immunosenescence. This process is not a simple decline but a complex remodeling of immune function that affects both innate and adaptive immunity. Key aspects of immunosenescence include:

  • Decreased T cell receptor (TCR) diversity: This limits the immune system's ability to recognize new antigens, which can hinder the response to both infections and cancer.
  • Reduced naïve T cell pool: The thymus, responsible for producing new T cells, atrophies with age, leading to a smaller pool of naïve, or inexperienced, T cells.
  • Chronic low-grade inflammation (Inflammaging): Aging is often accompanied by persistent, low-level systemic inflammation, which can affect the tumor microenvironment and immune responses.
  • Immune cell exhaustion: Chronic antigen stimulation can lead to T cell exhaustion, characterized by reduced proliferative capacity and increased expression of immune checkpoint molecules.

These age-related shifts in the immune system set the stage for how checkpoint inhibitors function in older patients, potentially affecting biomarker signaling and therapeutic outcomes.

Age-Related Changes in Key Biomarkers

The aging process directly alters the expression and function of several immune checkpoint therapy biomarkers, leading to a different prognostic picture in older adults.

Programmed Death-Ligand 1 (PD-L1)

  • Increased expression: Studies have shown that PD-L1 expression increases in senescent cells and aged tissues, as well as on tumor and stromal cells in elderly patients. This heightened expression could be a mechanism for senescent cells to evade immune clearance.
  • Implications for therapy: Higher PD-L1 levels are generally linked to a better response to checkpoint inhibitors. However, the increased PD-L1 on senescent immune cells rather than just tumor cells adds nuance to this biomarker's predictive power in older patients.

Tumor Mutational Burden (TMB)

  • Elevated with age: Research has linked increasing age with a higher tumor mutational burden in many cancer types.
  • Therapeutic advantage: A high TMB is often associated with a better response to immune checkpoint blockade. This may be one reason why some older patients show robust responses to therapy, potentially counteracting some of the negative effects of immunosenescence.

T-cell Receptor (TCR) Diversity

  • Decreased with age: As the immune system ages, the diversity of the T cell receptor repertoire diminishes. This can limit the variety of neoantigens the immune system can recognize and attack.
  • Implications for therapy: Reduced TCR diversity might lessen the effectiveness of ICB therapies by restricting the pool of T cells available to target tumor cells.

Clinical Outcomes: Older vs. Younger Patients

Clinical data on the effectiveness of immune checkpoint inhibitors in older versus younger patients has yielded mixed results, highlighting the complexity of age as a factor.

  • Some meta-analyses suggest comparable overall survival and progression-free survival between younger (<65) and older (≥65) patients receiving ICIs.
  • However, other studies indicate that the benefit may be smaller for very elderly patients (e.g., >75 years).
  • A study by Johns Hopkins found that older adults responded well to immunotherapy, despite differences in immune system response. Their analysis suggested unique immune responses in older patients that could be leveraged for future personalized therapies.
  • Factors such as frailty and comorbidities in older patients can impact outcomes, sometimes independent of the direct immunological efficacy of the treatment.

The Role of the Tumor Microenvironment

Age-related changes in the tumor microenvironment (TME) are also critical. The aged TME can be more immunosuppressive due to an increase in certain cell types and secreted factors. For example, the accumulation of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) in older individuals can suppress anti-tumor immunity. This creates a complex signaling network that immune checkpoint inhibitors must overcome to be effective. For example, some studies suggest that senescent-induced inflammation drives cancer growth by promoting pro-tumor macrophage accumulation.

Comparison of Immunotherapy Response in Different Age Groups

Factor Younger Patients (<65) Older Patients (≥65)
Immune System Robust T cell function, higher naïve T cell count, broader TCR diversity Immunosenescence, increased T cell exhaustion, smaller naïve T cell pool, restricted TCR diversity
Biomarker Profile Generally lower baseline levels of some inhibitory checkpoints Increased expression of immune checkpoints (e.g., PD-L1, LAG-3, TIM-3) on immune and senescent cells
Therapeutic Efficacy Consistent responses, but with potential for different irAE profiles Variable responses; some studies show similar or better outcomes, while others suggest reduced benefit over 75
Adverse Events Different irAE patterns, potentially more severe endocrine toxicity Different irAE patterns, potentially higher risk of certain irAEs, more frequent skin toxicities
Co-morbidities Typically fewer confounding health issues Higher prevalence of co-morbidities affecting treatment tolerance and outcomes

Future Directions and Personalized Care

Understanding how age affects immune checkpoint therapy biomarkers is a dynamic field of research with significant implications for patient care. Moving forward, a one-size-fits-all approach is insufficient. Geriatric-specific assessments that go beyond chronological age are needed to better stratify older patients and predict treatment tolerance and effectiveness. Additionally, novel therapeutic strategies, such as combining checkpoint inhibitors with senolytic drugs to clear senescent cells, are being explored to improve outcomes in older patients. Advancements in AI tools and comprehensive biomarker analysis promise to help guide personalized therapy selection.

Conclusion

Age is a critical factor influencing the efficacy of immune checkpoint therapy by altering predictive biomarkers and the overall immune landscape. The process of immunosenescence, marked by changes in T cell populations, inflammation, and checkpoint molecule expression, creates a unique context for immunotherapy in older adults. While clinical evidence shows comparable efficacy in many cases, individualized treatment approaches based on a deeper understanding of these age-related biological shifts are essential for optimizing care and improving outcomes for the growing senior population facing cancer. Further research is necessary to fully decipher the intricate interplay between aging, biomarkers, and immunotherapy response. https://www.nature.com/articles/s41467-025-58512-z

Frequently Asked Questions

Studies show that PD-L1 expression can increase in senescent cells and aged tissues, including on immune cells and some tumor cells. This upregulation is influenced by chronic inflammation and other age-related changes and can be a significant factor in immunotherapy response.

The evidence is mixed. While some studies suggest comparable efficacy between older and younger patients, others indicate that the benefits may be smaller for the very elderly (over 75). Factors like frailty and comorbidities often influence outcomes more than age alone.

Immunosenescence is the age-related decline and remodeling of the immune system. It affects biomarkers by altering T cell function, reducing receptor diversity, and increasing the expression of inhibitory checkpoints, ultimately influencing how a patient's body responds to immunotherapy.

A high TMB is generally a positive predictor for immune checkpoint inhibitor response, and TMB tends to increase with age. This may help explain why some older patients respond effectively to these therapies, as the higher mutation load creates more targets for the immune system to recognize.

Yes, research suggests that the types of immune-related adverse events (irAEs) can differ by age. For instance, skin toxicities may be more common in older patients, while endocrine toxicities might be more prevalent in younger patients.

Personalized care is crucial because an older patient's biological age and health status, including comorbidities and frailty, are often more predictive of treatment tolerance and outcome than their chronological age. Geriatric-specific assessments help tailor treatment plans accordingly.

Emerging research is exploring whether combining immune checkpoint inhibitors with other agents, such as senolytic drugs that clear senescent cells, could enhance therapeutic efficacy and restore immune function in older adults.

References

  1. 1
  2. 2
  3. 3
  4. 4
  5. 5
  6. 6
  7. 7

Medical Disclaimer

This content is for informational purposes only and should not replace professional medical advice. Always consult a qualified healthcare provider regarding personal health decisions.