Skip to content
RECEIVE FREE EXPRESS SHIPPING ON ALL ORDERS OVER $150 DELIVERED IN AUSTRALIA
RECEIVE FREE EXPRESS SHIPPING ON ALL ORDERS OVER $150 DELIVERED IN AUSTRALIA
A Naturopathic Perspective on PD-L1 Inhibitors

A Naturopathic Perspective on PD-L1 Inhibitors

Cancer immunotherapy has revolutionised how we approach cancer treatment, offering new hope to patients who previously had limited options. As a naturopathic practitioner, I'm particularly fascinated by PD-L1 inhibitors like Durvalumab, Atezolizumab, and Pembrolizumab medications that essentially flip the immune system's "on switch" for attacking cancer.

What makes these treatments so remarkable is that they don't create new immune responses or "build" immune cells from scratch. Instead, they release existing brakes on the immune system, freeing our natural cancer-fighting cells to do their job effectively.

"The most powerful medicine often doesn't add something new to the body, but rather removes the obstacles preventing the body from healing itself. PD-L1 inhibitors exemplify this principle perfectly."


In this post, I'll guide you through how these medications work from an immunological standpoint and explore several factors that influence their effectiveness - from gut health and chronic inflammation to bone marrow function. More importantly, I'll share practical, evidence-based strategies you can implement alongside conventional treatment to support your body's innate healing capacity.


Understanding the On/Off Switch in the Immune System

When functioning optimally, our immune system can recognise and eliminate cancer cells before they form tumours. However, cancer cells often evolve mechanisms to evade detection or suppress immune responses.

One of the most clever evasion tactics involves exploiting the PD-1/PD-L1 pathway - a natural "checkpoint" that normally prevents excessive immune activation. PD-1 (Programmed Death-1) is a receptor found on immune cells like T lymphocytes and some Natural Killer cells. Its partner, PD-L1 (Programmed Death-Ligand 1), can be displayed on other cells throughout the body.


When PD-L1 binds to PD-1, it essentially tells the immune cell to "stand down" - a critical mechanism for preventing autoimmunity under normal circumstances. However, many tumours have learned to express high levels of PD-L1 on their surface as a protective shield. When your body's T cells approach the cancer, this binding tricks them into hitting the brakes instead of attacking.


PD-L1 inhibitor drugs like Durvalumab, Atezolizumab, and Pembrolizumab  work by blocking this interaction. They bind to PD-L1 on cancer cells, preventing them from engaging with PD-1 on your T cells. This effectively disables the brake pedal, allowing your immune engine to rev at full power against the cancer.


The Cells That Do the Fighting
When we remove these immunological brakes with PD-L1 inhibitors, which cells actually do the cancer-fighting work? The stars of the show are CD8+ cytotoxic T lymphocytes (often called killer T cells) and, to a lesser extent, Natural Killer (NK) cells.


CD8+ T cells are sophisticated assassins that patrol your body looking for abnormal cells. When they recognise a cancer cell as a target, they form a tight bond and release toxic substances that punch holes in the cancer cell membrane and induce programmed cell death.Natural Killer cells serve as your immune system's surveillance team, eliminating stressed, virus-infected, or abnormal cells without needing a specific target. Both cell types can be suppressed by the PD-1/PD-L1 pathway, and both can be reinvigorated when we block this interaction.

"Cancer treatment isn't just about attacking the tumour - it's about cultivating an internal environment where your immune system can thrive. Think of PD-L1 inhibitors as removing the handcuffs from your body's natural cancer-fighting force."


Interestingly, the quality and quantity of these immune cells can vary significantly between patients, which helps explain why two people receiving the same PD-L1 inhibitor can have very different outcomes. This is where naturopathic and integrative approaches become particularly valuable - by optimising the health of these immune cells, we may enhance treatment efficacy.


Gut Health
One of the most fascinating developments in cancer immunotherapy research is the discovery that gut bacteria can significantly influence treatment outcomes. Patients who respond well to PD-1/PD-L1 inhibitors often have a more diverse and balanced gut microbiome compared to non-responders. Research has identified several bacterial species associated with better immunotherapy responses:


Akkermansia muciniphila: This mucin-degrading bacterium has been linked to better responses to PD-1 inhibitors in melanoma and lung cancer patients. It appears to enhance dendritic cell function and T cell recruitment to tumours.
Bifidobacterium species: Particularly B. longum, B. breve, and B. adolescentis have shown promise in enhancing anti-tumour immunity. These organisms produce short-chain fatty acids that regulate T cell function and reduce intestinal inflammation.
Faecalibacterium prausnitzii: A butyrate-producing bacterium that modulates regulatory T cells and reduces systemic inflammation.
Enterococcus hirae and Barnesiella intestinihominis: These have been associated with improved PD-1 inhibitor efficacy in both animal models and human studies.

In my practice, I recommend a fiber-rich, plant-diverse diet as the foundation for gut health. Aim for 30+ different plant foods weekly, including a variety of:
Vegetables and fruits in all colors
Legumes (beans, lentils, chickpeas)
Whole grains (if tolerated)
Nuts and seeds
Fermented foods like sauerkraut, kimchi, and yogurt (if dairy is tolerated)


For patients with compromised gut function, I sometimes recommend specific probiotic formulations containing these beneficial strains, but this must be individualised based on the patient's condition and concurrent treatments.


Immune Suppressors: Addressing MDSCs
Even with checkpoint inhibitors on board, your immune system may still face obstacles from cells called Myeloid-Derived Suppressor Cells (MDSCs). These immature myeloid cells accumulate in the blood and tumour environment of many cancer patients and act as powerful suppressors of immunity.
MDSCs employ various mechanisms to dampen immune responses: they deplete essential nutrients that T cells need to function, generate harmful reactive oxygen species, and secrete anti-inflammatory cytokines that convert T cells from killers to regulators. Essentially, they create an immunosuppressive shield around the tumour.


Nutraceuticals That May Reduce MDSC Activity
Several natural compounds have shown promise in modulating MDSCs in preclinical studies:
Curcumin: Found in turmeric, curcumin has been shown to reduce MDSC expansion and function through inhibition of STAT3 signaling. 
EGCG from green tea: Epigallocatechin gallate can reduce MDSC accumulation and activity. Consider 3-4 cups of organic green tea daily or 300-400mg of a standardised supplement.
Resveratrol: Found in grapes and berries, resveratrol has demonstrated the ability to reduce MDSCs in tumor-bearing mice. Therapeutic doses typically range from 500-2000mg daily.
Vitamin D: Maintaining optimal vitamin D levels (100-120 nmol/l) may help immature myeloid cells differentiate properly rather than becoming suppressive MDSCs.
All-trans retinoic acid (vitamin A): This vitamin A derivative can promote MDSC maturation into non-suppressive cells.

It's worth noting that these approaches should be discussed with your oncology team, as timing and dosing may need to be adjusted around treatment schedules.


Chronic Inflammation

Chronic inflammation creates a paradoxical situation for cancer patients - while acute inflammation is necessary for effective immune responses, chronic, smouldering inflammation often leads to immune dysfunction and exhaustion.
Two key players in this process are Interleukin-6 (IL-6) and the NF-κB pathway. IL-6 is commonly elevated in cancer patients with systemic inflammation and promotes the expansion of MDSCs and regulatory immune profiles that inhibit anti-tumour immunity. Similarly, chronic activation of NF-κB drives cells to secrete inflammatory cytokines that can support tumour growth and cause T cell exhaustion.


"Think of inflammation like a fire - a controlled burn can be beneficial and necessary, but a wildfire damages everything in its path, including your immune system's ability to target cancer."


 Evidence-Based Anti-Inflammatory Approaches
Here are some strategies I recommend to my patients to help manage chronic inflammation:
Omega-3 fatty acids: EPA and DHA from fish oil can reduce inflammatory cytokines and may improve immunotherapy outcomes. 
Specialised pro-resolving mediators (SPMs): These lipid mediators derived from omega-3s actively resolve inflammation rather than just suppressing it. 
Polyphenol-rich foods: Berries, cherries, pomegranates, and dark chocolate contain polyphenols that inhibit NF-κB activation. Include these foods regularly in your diet.
Modified citrus pectin: This specialised form of pectin has been shown to reduce galectin-3, a pro-inflammatory protein often elevated in cancer. Typical doses range from 5-15g daily.
Medicinal mushrooms: Reishi, turkey tail, and coriolus mushrooms contain compounds that can modulate inflammation while supporting immune function. Look for hot-water extracted products with verified beta-glucan content.

Time-restricted eating: Limiting eating to an 8-10 hour window daily may reduce inflammatory markers and improve metabolic health.


Remember that managing inflammation is a comprehensive approach that includes diet, stress management, sleep optimisation, and appropriate physical activity - all tailored to your individual needs and treatment plan.


Supporting Your Bone Marrow
Your bone marrow serves as the factory for new immune cells. For PD-L1 inhibitors to work optimally, you need a healthy repertoire of immune cells available to be activated.
Many cancer patients have compromised bone marrow function due to prior treatments like chemotherapy or radiation. Others might have age-related decline in marrow output or nutrient deficiencies that impair cell production.


Nutrients and Herbs for Bone Marrow Support
Here are key nutrients and botanicals that support healthy hematopoiesis (blood cell formation):
Protein and amino acids: Ensure adequate protein intake (at least 1.2-1.5g/kg body weight daily) to provide building blocks for cell synthesis.
Iron, vitamin B12, and folate: These are essential for DNA synthesis and red blood cell production. Have levels checked regularly and supplement if needed.
Zinc: Critical for immune cell development and function. Aim for 15-30mg daily from diet and supplements combined.
Astragalus (Astragalus membranaceus): This adaptogenic herb has been used traditionally to support bone marrow function and has shown promise in research for enhancing hematopoiesis after chemotherapy. 
Medicinal mushrooms: Specifically, Ganoderma lucidum (reishi) and Grifola frondosa (maitake) have demonstrated hematopoietic support in preclinical studies.
Chlorophyll-rich foods: Wheatgrass, chlorella, and spirulina provide chlorophyll, which has a molecular structure similar to hemoglobin and may support blood cell formation.
Remember that bone marrow function also depends on adequate rest, stress management, and avoiding toxins like cigarette smoke and excessive alcohol, which can directly damage hematopoietic stem cells.


Preventing T Cell Exhaustion
Even with ample supplies and removal of immunosuppressive factors, T cells can experience "burnout" from prolonged battle against cancer. T cell exhaustion is characterised by poor cytokine production, diminished killing ability, and the upregulation of multiple inhibitory receptors.
While PD-1/PD-L1 blockade can partially reverse exhaustion, giving these tired T cells a "second wind," the suppressive tumour microenvironment can still pose problems that one checkpoint inhibitor alone may not fix.


Supporting T Cell Function and Resilience
Here are strategies that may help maintain T cell vigor:
Optimise sleep: Deep sleep is when your body performs much immune regulation. Aim for 7-9 hours of quality sleep, using evidence-based sleep hygiene techniques.
Manage chronic infections: Controlling chronic viral infections (such as EBV or herpes viruses) can reduce background load on your immune system.
Strategic fasting: Emerging research suggests that short-term fasting or fasting-mimicking diets may enhance T cell function by improving metabolic flexibility and reducing glycolysis-dependent exhaustion. This approach should be supervised by healthcare providers familiar with both fasting protocols and cancer care.
Melatonin: Beyond its role in sleep, melatonin has immunomodulatory properties that may support T cell function. Doses of 3-20mg at bedtime may be beneficial for some patients (with medical supervision).
Coenzyme Q10: This mitochondrial supportive nutrient may help T cells maintain energy production. Consider 100-300mg daily of a highly bioavailable form.
N-acetylcysteine (NAC): This precursor to glutathione may help T cells maintain redox balance. Typical therapeutic doses range from 600-1800mg daily.

"The goal of integrated oncology isn't to replace conventional treatment, but to optimize your body's ability to benefit from it while minimising side effects. It's about creating the internal terrain where healing becomes possible."

PD-L1 inhibitors like Durvalumab represent a paradigm shift in cancer medicine - they work not by directly killing cancer cells, but by empowering our immune system to do so. This shift offers us a tremendous opportunity to participate actively in our healing process.
As patients or practitioners, we are not passive bystanders in this journey. By understanding the science behind these breakthrough therapies and addressing the factors that influence their effectiveness, we can become empowered partners in the healing process.
Think of it this way: the PD-L1 inhibitor is the general issuing orders, but the army is your own immune cells. Your lifestyle, nutrition, and self-care are the supply lines, training camps, and armoury for that army.
By incorporating the evidence-based approaches outlined in this article - supporting gut health, reducing chronic inflammation, nurturing bone marrow function, and preventing T cell exhaustion - you create an internal environment where immunotherapy can work most effectively.


References
Raskov H, Orhan A, et al. Cytotoxic CD8+ T cells in cancer and cancer immunotherapy. Br J Cancer. 2021;124(2):359–367.
Gazzaniga FS, Kasper DL. The gut microbiome and cancer response to immune checkpoint inhibitors. J Clin Invest. 2025;135(3).
Gopalakrishnan V, Spencer CN, et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103.
Hou A, Hou K, Huang Q, Lei Y, et al. Targeting myeloid-derived suppressor cells: a promising strategy to overcome resistance to immune checkpoint inhibitors. Front Immunol. 2020;11:783.
Bossi P, Trama A, et al. Baseline values of circulating IL-6 and TGF-β identify patients with HNSCC who do not benefit from nivolumab. Cancers (Basel). 2023;15(21):5257.
Méndez-López LF, González-Llerena JL, Vázquez-Rodríguez JA. Dietary Modulation of the Immune System. Nutrients. 2024;16(24):4363.
Oyer JL, Gitto SB, Altomare DA, Copik AJ. PD-L1 blockade enhances anti-tumor efficacy of natural killer cells. OncoImmunology. 2018;7(11).
Ahn E, Kissick HT, Ahmed R. Reinvigorating exhausted T cells by blockade of the PD-1 pathway. For Immunopathol Dis Therap. 2018;7(1-2):7–21.
Wen Y, Zhu Y, Zhang C, et al. Chronic inflammation, cancer development and immunotherapy. Front Pharmacol. 2022;13:1040163.
Gabrilovich DI, Nagaraj S. Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol. 2009;9(3):162–174.
Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91-97.
Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104-108.
Kim HR, Park JS, Park JS, et al. Gut microbiota and butyrate level changes associated with the long-term efficacy of immune checkpoint inhibitors in advanced non-small cell lung cancer. Oncoimmunology. 2021;10(1):1873530.
Yang F, Shi K, Jia YP, et al. Advanced glycation end products induce M1 polarization of macrophages through RAGE-mediated signaling pathway. Biochem Biophys Res Commun. 2020;531(1):37-44.
Nair S, Dhodapkar MV. Natural killer T cells in cancer immunotherapy. Front Immunol. 2017;8:1178.
Wesolowski R, Markowitz J, Carson WE. Myeloid derived suppressor cells – a new therapeutic target in the treatment of cancer. J Immunother Cancer. 2013;1:10.
Yu J, Wang Y, Yan F, et al. Noncanonical NF-κB activation mediates STAT3-stimulated IDO upregulation in myeloid-derived suppressor cells in breast cancer. J Immunol. 2014;193(5):2574-2586.
Suzuki E, Kapoor V, Jassar AS, et al. Gemcitabine selectively eliminates splenic Gr-1+/CD11b+ myeloid suppressor cells in tumor-bearing animals and enhances antitumor immune activity. Clin Cancer Res. 2005;11(18):6713-6721.
Nefedova Y, Fishman M, Sherman S, et al. Mechanism of all-trans retinoic acid effect on tumor-associated myeloid-derived suppressor cells. Cancer Res. 2007;67(22):11021-11028.
Trikha P, Carson WE. Signaling pathways involved in MDSC regulation. Biochim Biophys Acta. 2014;1846(1):55-65.

Disclaimer: This article is for informational purposes only and does not constitute medical advice. Always consult with your healthcare providers before making changes to your treatment plan or adding supplements, especially during active cancer treatment.

 

Next article The Molecular Science Behind Anti-Inflammatory Eating: Why Your Diet Is Your Most Powerful Medicine