Header image

SYMPOSIUM 10: Food-drug interaction in oncology: Implications for dosing, toxicity and economics of care

Tracks
Track 2
Tuesday, September 23, 2025
10:30 AM - 12:00 PM
Grand Copthorne Waterfront Hotel - Waterfront Ballroom I

Details

The TDM in Oncology sub-committee is pleased to propose a symposium on " Food-drug interaction in oncology: Implications for dosing, toxicity and economics of care". Food-drug interaction are perhaps the earliest documented interactions but least appreciated. Cancer drugs like irinotecan, tamoxifen, and paclitaxel exhibit significant variability in pharmacokinetics among different patients, influenced by genetic, physiological, and environmental factors, including diet. Besides, majority of these drugs typically have a narrow therapeutic index. Food is perhaps the only factor that varies significantly across ethnicities in terms of protein, fat and micronutrient content. Besides, the unique nutritional supplement consumption practices across populations can potentially exacerbate the variability of cancer drug exposure and consequently its safety and efficacy. Foods that modulate drug-metabolizing enzymes (eg. grapefruit) or affect drug absorption (high-fiber foods), can lead to unpredictable plasma levels of cancer drugs. Additionally, pharmacodynamic interactions (eg. catechins in green tea inhibit the action of bortezomib) may impact drug response. This symposium would enable oncologists and clinical pharmacologists to share evidence-based insights on such interactions. By addressing how food impacts drug PK-PD, this forum aims to promote safer, more effective cancer treatments that account for each patient's unique response, ultimately reducing adverse effects and improving therapeutic success.


Speaker

Agenda Item Image
Amy Rieborn
Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center

The pharmacological effects of using cabozantinib with a light breakfast: The SKIPPY1 study

Abstract

Introduction:
Cabozantinib is a tyrosine kinase inhibitor approved for treatment of renal cell carcinoma. Cabozantinib is started at 60 mg daily, ingested in fasted state. Dose reductions are needed in 67% of patients due to toxicity, without reduce in treatment costs due to a flat pricing model. Alternative dosing strategies are warranted to improve the toxicity profile and reduce treatment costs. Previous studies have shown cabozantinib AUC increased 57% after taking cabozantinib with a high fat meal. The increase in bioavailability when taken with a light breakfast is still unknown. In this study we aim to determine the increase of cabozantinib AUC when taken with a light breakfast.

Methods:
A multicentre, open label, randomized cross-over pharmacokinetic evaluation study was performed. In the standard regimen, patients took cabozantinib in fasted condition. In the experimental regimen, patients took cabozantinib with a light breakfast. After 4 weeks on the allocated regimen, pharmacokinetic samples were obtained at t=0,1,2,3,4,6,8 and 24 hours after dose intake and patients thereafter switched to the other regimen. Patients were monitored for (S)AEs.

Results:
Twelve patients completed study procedures. AUC0-24 for taking cabozantinib in fasted and fed state were 11530 ug/L and 12530 ug/L respectively (CV 10.1%). Mean difference was 8.3% (ratio 90% CI 100.9 – 117.12). No differences in adverse events were established.

Discussion and conclusion:
This study demonstrates taking cabozantinib with a commonly used breakfast leads to a slight increase in exposure, but with no apparent impact on adverse events. These results support to ingest cabozantinib with food without clinically relevant influence on exposure.

Biography

Amy Rieborn is a PhD candidate at the departments of Clinical Pharmacy and Toxicology and Medical Oncology of Leiden University Medical Center. Her research is focused on therapeutic drug monitoring (TDM) and personalized medicine in oncology. Trained as a medical doctor with previous work experience in the field of internal medicine, she aims to translate the theoretics of TDM and pharmacokinetic modelling into a direct benefit for the patient in daily practice.
Agenda Item Image
A/Prof Ashley Hopkins
Flinders University

Gastrointestinal Environment and Drug Interactions in Cancer Patients: Insights from Clinical Trial Data

Abstract

This presentation showcases research underpinned by data transparency frameworks that enabled negotiation of access to patient-level data from over 100 industry-sponsored oncology trials. These rich datasets have facilitated investigations into how gastrointestinal-modifying concomitant medicines influence survival and adverse event outcomes in cancer.
Key findings from Associate Professor Ashley Hopkins’ work demonstrate that proton pump inhibitor (PPI) use is associated with poorer survival benefit from immunotherapies, such as atezolizumab, in non-small cell lung cancer. Large-scale prognostic analyses further reveal that PPI exposure correlates with reduced overall survival and worsened toxicity in colorectal and breast cancer cohorts. Methodological approaches include multivariable Cox regression and pooled post hoc trial evaluations, illustrating how systematic analyses of participant-level data can uncover clinically meaningful drug interactions.
These results underscore the urgent need to advocate for and expand transparent access to trial data. Findings highlight that GI modulating drugs can alter the gut microbiota and gastric pH, affecting drug absorption, pharmacokinetics, and pharmacodynamics—factors of growing importance as oncology shifts toward orally administered targeted therapies.

Biography

Associate Professor Ashley Hopkins leads the Clinical Cancer Epidemiology Lab at Flinders University, South Australia. His research harnesses large-scale clinical trial data to understand how commonly used medications, including those affecting the gastrointestinal environment, influence cancer treatment outcomes. Through industry collaborations, he has negotiated access to individual participant data from over 100 oncology trials, enabling world-first analyses on the impact of antibiotics, antacids, and body composition on immunotherapy efficacy and toxicity. Ashley is a strong advocate for data transparency, contributing to international frameworks through roles with the Clinical Oncology Society of Australia and the Society for Immunotherapy of Cancer. His work has shaped clinical guidelines, featured in high-impact journals such as JAMA Oncology, Journal of Clinical Oncology, and The BMJ, and earned recognition through the Tall Poppy Award, Certara New Investigator Award, and prestigious fellowship grants. He is also committed to mentoring emerging researchers and embedding consumer perspectives in cancer research.
Agenda Item Image
Prof Jennifer Martin
School of Medicine and Public Health, The University of Newcastle

Effect of dietary proteins on the pharmacokinetics and toxicity of 5-flurouracil

Biography

Professor Jennifer Martin is a senior physician and clinical pharmacologist with 30 years clinical, Government, industry and research experience in optimising drug choice and dose. One aspect of this work is therapeutic drug monitoring to optimise individual dosing. Although the genetic makeup of some individuals explains some of the exposure, it doesn’t explain most of the variation in clinical exposure and outcomes. Recently, the effects of diet (which may be a surrogate for culture, geography or microbiome) are seen to contribute significant effects on clinical exposure, and/or clinical outcomes. This may be particularly relevant to the use of purine, pyrimidine and other metabolic analogues such as 5FU. This talk will highlight the literature in this space drawing from the Cancer Council funded 5FU dose optimisation studies in Newcastle.
Agenda Item Image
Prof Frank Jansman
Deventer Hospital and University Groningen

Clinical implications of food–drug interactions with small-molecule kinase inhibitors

Abstract

During the past decades, small-molecule kinase inhibitors have proven to be valuable in the treatment of solid and haematological tumours. However, because of their oral administration, the intrapatient and interpatient exposure to small-molecule kinase inhibitors (SMKIs) is highly variable and is affected by many factors, such as concomitant use of food and herbs. Food–drug interactions are capable of altering the systemic bioavailability and pharmacokinetics of these drugs. The most important mechanisms underlying food–drug interactions are gastrointestinal drug absorption and hepatic metabolism through cytochrome P450 isoenzymes. As food–drug interactions can lead to therapy failure or severe toxicity, knowledge of these interactions is essential.
Therefore, general principles involving food–drug interactions and herb–drug interactions for SMKIs will be presented. For illustration, the design and results of specific studies will be discussed more extensively. Also, US Food and Drug Administration (FDA) and European Medicines Agency (EMA) recommendations concerning food-drug interactions are considered. Based on current available information, clear recommendations for their management will be provided, and suggestions for performing food-drug studies during drug development phases and thereafter.

Biography

Frank Jansman is professor in clinical pharmacy, in particular oncology-pharmacy at the University Groningen. In addition, he is head and certified trainer hospital pharmacy of the department of clinical pharmacy of the Deventer Teaching Hospital. He combines his activities in the hospital with research and education for the Groningen Research Institute of Pharmacy and for the study pharmacy. Reseach lines include medication surveillance and pharmaceutical care for cancer patients and factors influencing anti-cancer drug exposure in patients. Given his chair of the Dutch Expertgroup Anticancer Drug Interactions, research to co-medication, alternative medicines, and food in combination with anti-cancer drugs is a major topic. Frank Jansman is a hospital pharmacist and a clinical pharmacologist.
Agenda Item Image
Dr Vikram Gota
Advanced Centre For Treatment, Research and Education in Cancer, Tata Memorial Centre, Mumbai

Food effect on the bioavailability of abiraterone and its implications for dosing and mitigating financial toxicity: Results of a randomized controlled trial

Abstract

Abiraterone acetate is a cornerstone in the management of metastatic castration-resistant prostate cancer (mCRPC). Given its food effect on absorption, we compared outcomes between a low-dose (250 mg daily- Arm A) and standard-dose (1000 mg daily – Arm B) abiraterone regimen. The low-dose abiraterone was administered with a low-fat meal, whereas the standard dose was administered in fasting state as per standard recommendations. The objective of the study was to assess whether lower dosing could maintain efficacy and safety while improving cost-effectiveness. In this study, overall survival (OS) and median prostate-specific antigen progression-free survival (PSA-PFS) were comparable between the two arms [PSA-PFA = 5.7 vs 3.8 months, HR=0.95; OS = 18.1 vs 15.1 months, HR=0.99]. Similarly, the incidence of grade ≥3 adverse events did not differ significantly [36.6% vs 31.6%, p=0.51]. Pharmacokinetic (PK) analysis showed that both area under the concentration-time curve (AUC) and maximum plasma concentration (Cmax) were lower in the low-dose arm compared to the standard-dose arm [AUC = 28.7 vs 226 ng/ml*h], indicating reduced systemic exposure. Importantly, the anticipated improvement in bioavailability with a low-fat diet was not observed in the low-dose group. Despite lower PK exposure, clinical outcomes and toxicity remained similar between low-dose and standard-dose treatments. These findings suggest that administering low-dose abiraterone with a low-fat meal does not enhance bioavailability but achieves comparable survival and safety outcomes to the standard regimen. This approach could represent a meaningful strategy for substantial cost savings without compromising patient care.

Biography

Dr. Vikram Gota completed his MD in pharmacology from Christian Medical College, Vellore, following which he briefly worked as a Clinical Investigator in BA/BE studies. Thereafter, he joined the INDO￾Oxford (INDOX) Cancer Trials Network at Tata Memorial Centre, Mumbai, where he received training in the design and conduct of phase I clinical trials. During this time, he also obtained the post graduate diploma in clinical trials from the London School of Hygiene and Tropical Medicine, University of London. He is presently the officer-in-charge of the department of clinical pharmacology at the Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre. His research interests include early clinical development and pharmacokinetics driven optimization of drugs used in cancer, including the development of pediatric-friendly formulations for childhood cancers. He is currently the Chair of the TDM in Oncology sub-committee of the International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT). He is involved with the geriatric clinic at TMH since 2019 focusing on rational therapeutics, potentially inappropriate medications and drug-drug interactions, among other things. He has worked as an investigator in several phase I clinical trials including first-in-human studies of investigational new drugs.

Session chair

Agenda Item Image
Vikram Gota
Advanced Centre For Treatment, Research and Education in Cancer, Tata Memorial Centre, Mumbai

Ganessan Kichenadasse
Flinders University

loading