Abstract
Energy balance accounts for an individual’s energy intake, expenditure, and storage. Each aspect of energy balance has implications for the pharmacokinetics of cancer treatments and may impact an individual’s drug exposure and subsequently its tolerance and efficacy. However, the integrated effects of diet, physical activity, and body composition on drug absorption, metabolism, distribution, and excretion are not yet fully understood. This review examines the existing literature on energy balance, specifically the role of dietary intake and nutritional status, physical activity and energy expenditure, and body composition on the pharmacokinetics of cancer therapeutics. As energy balance and pharmacokinetic factors can be influenced by age-related states of metabolism and comorbidities, this review also explores the age-related impact of body composition and physiologic changes on pharmacokinetics among pediatric and older adult populations with cancer.
In 2022, there will be approximately 2 million new cancer cases and more than half a million cancer deaths in the United States alone (1). Although enormous strides have been made in cancer treatments over the last several decades, variability in treatment tolerability, particularly for chemotherapy, remains an important issue in clinical practice. Given the narrow therapeutic range of many cancer therapies, understanding what impacts individual patient variation in drug exposure and treatment tolerability is critical to optimizing care. Understanding how medications move throughout the body, including their absorption, metabolism, distribution, and excretion, known as pharmacokinetics (PK), is paramount to optimizing treatment decisions and dosing considerations (2).
One crucial factor that may contribute to the PK of cancer therapies is energy balance, which consists of energy (dietary) intake, energy expenditure, and energy storage. There is growing evidence that specific aspects of energy balance, such as healthy dietary intake and moderate to high levels of physical activity, can reduce the risk of morbidity and mortality after a cancer diagnosis. However, how energy intake and expenditure per se impact chemotherapy PK and treatment tolerability is less understood. Furthermore, the role of body composition as an index of energy storage in predicting cancer outcomes is of increasing interest within oncology (3,4). Low muscle mass and strength, commonly known as sarcopenia, are strongly associated with survival and chemotherapy toxicity (5,6). Similarly, sarcopenic obesity (eg, low muscle and excess fat mass) and myosteatosis (fat infiltration in muscle) are risk factors for increased toxicities and adverse outcomes (7,8). How these factors precisely influence PK and chemotherapy toxicities remains unclear.
The overall objective of this review is to examine the existing literature on the impact of energy balance on the PK of cancer therapeutics, particularly chemotherapy (Figure 1). More specifically, we examine the role of dietary intake, nutritional status, energy expenditure, and body composition in influencing cancer treatment PK. In addition, we explore the existing evidence of energy balance parameters impacting PK related to special populations, including pediatric and older adult populations with cancer. For this review, we focused on energy balance and PK given our interest in how the body and energy balance may affect cancer therapies. Pharmacodynamics (PD) is a related topic that examines how a drug affects the body and its intended biologic effect, but it is not a focus of this review.
Figure 1.
Impact of energy balance on pharmacokinetics of cancer therapeutics.
Energy intake: diet and nutrition
Acute dietary intake
Acute (recent) dietary intake can substantially impact the PK of some oral drugs, including many cancer therapeutics (9). Compared with fasted conditions, acute dietary intake can alter gastric acidity, increase splanchnic and hepatic blood flow, stimulate bile production and release, and influence gastrointestinal transit times (9,10). Collectively, these factors can impact absorption, distribution, metabolism, and excretion—and, hence, the PK—of oral drugs (11,12). Earlier work has demonstrated that half of all chemotherapeutics approved by the US Food and Drug Administration (FDA) between 1971 to 2013 required consumption either in fasted or fed conditions, with the other half not requiring consideration of prior food (13).
Because many drugs interact with food, food-effect bioavailability studies are a cornerstone for assessing the efficacy and tolerance of orally administered drugs and for obtaining FDA or European Medicine Agency approval for use (12,14). Currently, the FDA and European Medicine Agency recommend assessing oral drug bioavailability after consuming a meal high in energy and fat (800-1000 kcal; 500-600 kcal from fat). If a food–drug interaction is present, further testing with a meal containing a moderate amount of energy and fat (400-500 kcal; approximately 150 kcal from fat) may be warranted (14); however, there is no guidance regarding low-fat meals (15).
Guidelines on diet–drug interaction testing and directions for use are largely adequate for ensuring safe and effective chemotherapy dosing. However, there are specific foods or acute dietary intake patterns that may impact the PK of chemotherapeutics that are not identified in pre-approval testing. In one example of a randomized crossover trial, 28 patients with non-small cell lung cancer received concomitant treatment with erlotinib (targeted therapy) with or without esomeprazole (proton pump inhibitor) and with either 250 mL of cola or water for 7 days (16). In patients taking proton pump inhibitors, the mean area under the plasma concentration time curve (area under the curve [AUC]) was 39% higher, and the maximum serum concentration was 42% higher after consumption of the cola compared with water, likely because cola alters stomach acidity and thus the solubility and availability and PK of erlotinib (16). In another example, low-dose (250 mg) abiraterone acetate (androgen synthesis inhibitor) consumed with a low-fat meal was noninferior to the standard dose (1000 mg) consumed in fasting conditions among 36 patients with metastatic castration-resistant prostate cancer (17). Given the diversity of dietary intake in free-living conditions, continued exploration of acute diet–PK interactions could have substantial ramifications for patients, prescribers, and payers—especially for drugs with a narrow therapeutic range in which acute dietary intake could precipitate underdosing or toxicity (18). Notably, PK-informed recommendations regarding acute dietary intake often require behavior changes, which may not be followed by all patients. For example, 41% of individuals with cancer did not consider their last meal before taking oral chemotherapy, and only 20% correctly identified food–drug interactions from their chemotherapy (19). Recommendations for acute dietary intake also assume patients have adequate resources and knowledge to make appropriate changes (eg, the meaning of low fat or fasting). However, there is a lack of dissemination of trustworthy nutrition information for people with cancer (20). Cultural and religious practices such as Ramadan fasting may also impact dietary intake and thus PK of the chemotherapeutics used (21,22). Therefore, delineating diet–PK interactions may help refine behavioral approaches for enhancing cancer treatment efficacy.
Moderate- and long-term dietary intake
Whereas acute dietary intake is an important consideration in PK, there is limited knowledge on how moderate- or long-term dietary habits over days, weeks, or months may affect cancer therapeutics PK. Data from oral drugs used for conditions other than cancer suggest that some dietary patterns elicit changes in whole-body metabolism that impact the absorption, distribution, metabolism, and excretion of drugs. For example, changes in macronutrient distribution may alter drug-protein binding capacities, cholesterol levels, or bile acid turnover, which in turn impact serum concentrations of antiepileptic drugs (23,24). Even 3 days of a high-fat diet can increase the exposure of midazolam and omeprazole in healthy individuals, potentially through alterations in the cytochrome P450 enzyme system (25). Similarly, a 10-day high-protein, low-carbohydrate diet can increase the clearance of propranolol and theophylline in healthy individuals (26).
In the context of cancer, most studies of moderate- or long-term diet–PK interactions have been conducted in animals and generally report that protein or energy intake restriction results in lower clearance or increased absorption of common chemotherapeutic and other cancer agents (ie, 5-fluorouracil, methotrexate, anthracycline, vinca alkaloids, etoposide, erlotinib) (27). Animal studies can clarify the mechanistic basis for PK in response to dietary patterns in controlled settings. However, human applicability may be limited because animal studies often use extreme modulations in protein or energy intake implausible in humans. Many studies of chemotherapy agents in animals have administered diets devoid of protein or only approximately 3% of energy intake from protein, whereas protein malnutrition has been associated with altered clearance or toxicity profiles of chemotherapeutic agents (28,29).
Understanding dietary intake–PK relationships in humans is also vital, considering increased public and research interest in intermittent fasting, fasting-mimicking diets, or time-restricted eating practices to improve response to certain chemotherapies (30,31). It has been suggested that fasting may reduce the amount and severity of toxicities while preserving antitumor effects treatment (32); however, whether this occurs through changes in PK is largely unknown. In one published randomized crossover trial (and the only one of its kind to our knowledge), patients with solid tumors and liver metastases underwent 30% energy intake restriction and 70% protein restriction for 5 days before irinotecan administration vs their regular diet (33). Calorie and protein restriction resulted in higher 24-hour plasma AUC of irinotecan and its active metabolite SN-38 compared with regular diets, with no difference in toxicities. Notably, protein and calorie restriction resulted in weight loss over the first chemotherapy cycle, although most patients regained their initial weight 2 weeks after the diet period. As previously reviewed, fasting may be a potentially feasible and promising method of maximizing treatment efficacy; however, current evidence in humans is limited and primarily focuses on extended fasting protocols (34) (refs). Short-term fasting or intermittent fasting may be particularly relevant given the promise of chronomodulated chemotherapy in which treatment administration is based on circadian rhythms. As highlighted by a recent systematic review, this approach may reduce toxicity and preserve the efficacy of many common chemotherapy regimens (35). Interestingly, although many chronomodulated chemotherapy regimens included in the review scheduled dosing initiation or peak in the early morning hours, none considered recent dietary intake, dietary patterns, or fasting. Thus, the relative contribution of biological variation based on circadian rhythms or behavioral factors such as the timing of dietary intake on treatment outcomes is unclear. Given the potential positive effects of fasting but adverse effects of weight loss in some patients with cancer (36,37), more mechanistic PK research is needed to identify which patient populations may benefit from specific and novel dietary intake patterns.
Nutritional status
The National Cancer Institute defines nutritional status as “the state of a person’s health in terms of the nutrients in his or her diet.” It considers a more comprehensive view of health by encompassing patient characteristics such as anthropometric data (including body composition), biochemical parameters, clinical assessment, and dietary intake.
There are several methods to determine nutritional status, but the Scored Patient-Generated Subjective Global Assessment (PG-SGA) is one of the most commonly used tools in research and clinical oncology settings (38,39). This questionnaire is a 4-in-1 clinical instrument for nutritional screening, assessment, intervention triage, and monitoring. Several previous studies have found scores from the full and short forms of the PG-SGA predict important clinical outcomes such as length of hospital stays, quality of life, postoperative complications, medical costs, and survival (40). There is also evidence that malnutrition, as assessed by the PG-SGA, is related to a higher prevalence of treatment toxicities and dose reductions and poorer treatment response rates and disease control (41-43).
The predictive ability of the PG-SGA is likely because it measures global risk for malnutrition rather than nutritional deficit alone (40). However, it is not clear if associations between PG-SGA score and treatment outcomes are mainly due to alterations in physical function and body composition or if nutritional status as assessed by the PG-SGA directly relates to PK. As an illustrative example, we assessed PG-SGA scores and oxaliplatin PK among 25 older adults with gastrointestinal cancers [NCT03998202 (44)]. As shown in Figure 2, PG-SGA total score was negatively correlated with the volume of distribution and clearance of oxaliplatin, indicating that worse nutritional status was associated with lower values of these 2 PK parameters (and higher exposure to oxaliplatin). After adjusting for body surface areas via partial correlation, the relationship between PG-SGA and clearance remained statistically significant (Spearman rho = −0.419; P = .047), with a trend toward statistical significance between the PG-SGA and volume of distribution (Spearman rho = −0.397; P = .061). Given this exploratory data, altered PK may contribute to worse clinical outcomes observed in patients with poor nutritional status. However, the utility of using more general and accessible measures of long-term and current nutritional status—such as the PG-SGA—to predict PK-related parameters is unknown and needs to be established in future studies.
Figure 2.
Relationship between total short-form Patient-generated Subjective Global Assessment (PG-SGA) score and volume of distribution (A) and clearance (B) of oxaliplatin. Participants were n = 25 patients with gastrointestinal cancer (n = 12 colon, n = 7 pancreatic, n = 3 esophageal, n = 1 appendix, n = 1 rectal), with a median age of 69 years and predominantly male (68%).
Energy expenditure: focus on exercise
Total daily energy expenditure consists of resting energy expenditure (energy used to maintain bodily function; the largest component of total daily energy expenditure), physical activity energy expenditure (energy used to move the body), and the thermic effect of food (energy used to digest and assimilate consumed nutrients). In patients with advanced cancer, tumor burden and associated systemic inflammation may increase resting energy expenditure (45), but this may not be reflected in free-living total energy expenditure in patients with primarily earlier-stage cancer (46). Although preliminary evidence suggests that resting energy expenditure may relate to cancer treatment outcomes (47), no study to date has assessed energy expenditure and PK specifically. Similarly, there is limited data on the thermic effect of food in cancer in general (48-50), and none that investigates this energy expenditure component in relation to PK. As such, we will focus our discussion on energy expenditure–PK relationships on physical activity energy expenditure.
Physical activity energy expenditure is broadly categorized as the energy used to perform structured exercise or nonexercise activities (eg, movement from occupations, sitting, standing, ambulatory activities). Exercise is a critical consideration in the PK of many drugs because it acutely increases cardiac output, which enhances blood flow to metabolically active tissues and organs (eg, skeletal muscle, heart, lungs). Greater blood flow affects PK activity by increasing the amount of drug reaching appropriate receptor sites. Hence, as muscle blood flow increases with exercise, drug absorption and equilibrium between plasma and organ and/or tissues increases. When exercise ceases, constriction of arterioles in muscles and decreased splanchnic blood flow decrease drug re-entry in the circulatory system. Regular exercise can enhance the metabolic efficiency and activity of drugs, perhaps through physiological changes in cytochrome P-450 isozyme levels, plasma volume, plasma proteins (eg, serum albumin), body composition, capillarization of heart and skeletal muscle, and tissue mitochondrial density. Drug-specific factors such as membrane permeability and pH and whether a drug is flow limited (weakly bound, highly extracted) or capacity limited (strongly bound, poorly extracted) may impact exercise–PK relationships. However, the limited data in this area preclude definitive estimations of drug distribution by exercise.
Although much can be gleaned from the effect of exercise and the PK of drugs in the realm of cardiovascular disease and diabetes (eg, exercise increases the rate of insulin absorption in patients receiving basal dosing) (51,52), no studies to date have examined the role of exercise on the PK of cancer drugs. This research gap was recently highlighted by an attempt to perform a systematic review on this topic by Curnier (53); following an extensive literature search, the authors concluded there is a lack of data examining the role of exercise on the PK of cancer drugs. Theoretically, exercise influences on PK mentioned above may subsequently influence chemotherapy efficacy. Several preclinical models have concluded that exercise can affect cancer treatment efficacy by slowing tumor growth and progression (54-56). These outcomes have been attributed to regulating intratumoral vascular maturity, perfusion, hypoxia, and tumor cell metabolism (57), suggesting improved drug delivery and a positive treatment effect.
Exercise increases the efficiency of oxygen utilization by muscle and thus increases aerobic capacity. Aerobic capacity, or VO2 max, is the maximum volume of oxygen the body can utilize in a given amount of time. By increasing aerobic capacity and producing greater uptake and delivery of oxygen during exercise, the typically hypoxic tumor microenvironment may be reversed (58). One mechanism for this is the effect of exercise on tumor vasculature through its remodeling of unorganized vessel sprouts to more organized and elongated vessels that allow for increased oxygen delivery to the tumor, as well as increased drug delivery and distribution (59). Studies like this have laid the groundwork to support safety and feasibility studies of exercise during chemotherapy infusion (60); however, alterations in cancer drug metabolism have yet to be studied.
Clinical studies have also demonstrated that exercise improves cancer treatment–related side effects, which may lead to improved treatment completion rates (61). This decrease in side effects may be related to multiple PK-related effects of exercise on chemotherapy that have yet to be studied. For example, exercise may decrease unwanted side effects of chemotherapy by increasing the rate of excretion or changing the distribution of the drug. It may stand to reason that an increased excretion rate may negatively impact chemotherapy efficacy, yet preclinical models have shown the opposite through a synergistic effect with chemotherapy (56). In multiple mouse models of sarcoma tumors, when exercise was combined with chemotherapy, a greater reduction in tumor burden was seen when compared with chemotherapy alone, thus synergistically improving chemotherapy efficacy (56,59). Despite preliminary evidence suggesting exercise improves treatment adherence and rate of completion, future clinical studies on the effect of exercise on the bioavailability of cancer drugs are necessary to understand whether these changes impact desired therapeutic outcomes.
Regardless of PK, the latest American Society of Clinical Oncology guidelines on exercise, diet, and weight management during cancer treatment strongly recommend aerobic and resistance exercise during active cancer treatment to mitigate side effects (62). Although clinical evidence currently does not sufficiently support exercise to improve cancer control or improvement in relative dose intensity of therapy regimens, the physical and psychosocial benefits outweigh the harms. Benefits may include reduced fatigue; preserved cardiorespiratory fitness and physical function and strength; and improved quality of life, anxiety, and depression with a low risk of adverse events. Further research is needed to understand if altered PK that maximizes chemotherapy treatment is also a benefit of exercise.
Body composition
With the growing ability to utilize routine computed tomography (63) for body composition analyses, many studies have examined the link between body composition and adverse outcomes in oncology (4). The first meta-analysis of sarcopenia demonstrated a 44% increased risk of death in patients with cancer and low skeletal muscle mass, regardless of cancer type or disease stage (5). Similarly, low skeletal muscle mass is predictive of 4.08 higher odds of severe chemotherapy toxicity (95% confidence interval [CI] = 2.48 to 6.70) and 2.24 higher odds of dose-limited chemotherapy toxicity (95% CI = 1.28 to 3.92) (64). What underpins these associations remains unclear and is a focus of ongoing research. In 2016 Prior et al. (65) found that sarcopenia is associated with lower exercise capacity, which could result in limiting diffusion of substrates, oxygen, hormones, and nutrients necessary to counter metabolic-related chemotherapy toxicities. Many initially hypothesized that low skeletal muscle mass was synonymous with reduced performance status, physical function deficits, and increased frailty; however, these deficits are poorly associated with skeletal muscle mass (66,67). An alternative hypothesis increasingly gaining traction to explain the increased chemotherapy toxicities is related to altered PK. The dosing of the vast majority of cytotoxic chemotherapeutics is based on body surface area (BSA). BSA was initially proposed to normalize chemotherapy dosing because of the proposed relationship of BSA with other physiologic parameters, including resting energy expenditure, plasma volume, and cardiac output (68). Although several studies have shown that BSA only modestly reduces interpatient drug variability (69,70), it remains commonplace in oncology practice. BSA-based dosing relies solely on height and weight and does not consider variations in body composition (71). A wide distribution of lean body mass and adipose is observed for any given BSA and may affect altered chemotherapy volume of distribution, metabolism, and clearance of hydrophilic and lipophilic drugs from the systemic circulation thereby reducing treatment efficacy and excess toxicity.
In contrast to the many studies examining body composition and cancer outcomes, only a few studies have examined body composition and drug PK (72). In one of the first studies in 2011 by Prado et al. (73), lean body mass was highly variable, associated with toxicities, and explained 33% of the variance in the clearance of epirubicin in patients with breast cancer. Importantly, BSA was not predictive of lean body mass or associated with epirubicin clearance (73). In 2012 Mir et al. (74) found similar results with sorafenib in patients with advanced hepatocellular carcinoma. Patients with sarcopenia experienced significantly more dose-limiting toxicities (82% vs 31%; P = .005) and had statistically significantly higher median sorafenib plasma AUC (102.4 mg/l.h vs 53.7 mg/l.h; P = .013). A more recent study examined oxaliplatin PK in 26 older adults with gastrointestinal malignancies and showed the body composition phenotype of low lean body mass and high total adipose tissue had the lowest volume of distribution, lowest clearance, highest max drug concentrations, and a 45% increased risk of severe grade 3-5 chemotherapy toxicities (2). Population PK models of oxaliplatin demonstrated that total adipose tissue, lean body mass, and serum albumin explained 14%, 11%, and 17%, respectively, of oxaliplatin variability beyond BSA.
However, not all studies have observed similar findings. The results for 5-fluorouracil have been mixed, with one study finding no association between skeletal muscle mass and first-cycle AUC. In contrast, another study found improvements in the 5-fluorouracil clearance model fit with the addition of the skeletal muscle index (75,76). Another study of capecitabine, an oral prodrug of 5-fluorouracil, found no association between skeletal muscle mass and PK parameters (77). These contrasting findings are not wholly unexpected, as the association between chemotherapy PK and body composition will largely be drug specific and depends greatly on the hydrophilic or hydrophobic nature of the specific compound and how it distributes within the body.
Special populations
Pediatrics
Because of major advances in cancer-directed therapies over the last several decades, long-term survival for childhood cancer is now more than 80% (78). However, owing to their exposure to antineoplastic agents, childhood cancer survivors experience a significantly greater burden of chronic health conditions at a far younger age when compared with the general population (63). Gaining a better understanding of energy balance and chemotherapeutic PK in children as they age into adolescence and young adulthood may enable a more precise mechanism of dosing antitumor agents in the pediatric population, thereby potentially mitigating some of the short- and long-term adverse effects in this at-risk group (79).
To our knowledge, no study has assessed dietary intake and exercise on PK parameters in pediatrics per se. However, some have investigated the “energy storage” aspect of energy balance by characterizing body weight or composition on PK (79). Though the poor association between body mass index (BMI) and body composition is well recognized, and it is understood that traditional methods of dosing by BSA do not accurately predict chemotherapy PK, pediatric clinical studies involving chemotherapy PK have primarily focused on the effects of BMI (80). Results of these investigations have been mixed; however, publications on conventional chemotherapy agents including doxorubicin, mercaptopurine, busulfan, and more recently, high-dose methotrexate and asparaginase demonstrate that adiposity does alter cytotoxic drug activity among children with cancer (81-84).
Browning and colleagues (81) demonstrated that weight-based dosing of busulfan among children with high BMI resulted in higher than expected drug concentrations. Conversely, Zuccaro et al. (82) showed that dosing of 6-mercaptopurine based on BSA resulted in underdosing among overweight children. Thompson et al. (83) showed that pediatric oncology patients with more than 30% body fat exhibited disturbed clearance of the doxorubicin metabolite, doxorubicinol. Similarly, Orgel and colleagues (84) showed that, among acute lymphocytic leukemia patients, obesity was associated with delayed clearance of high-dose methotrexate and increased risk of asparaginase-associated toxicity. Human studies align with some preclinical studies, showing that vincristine PK is altered in obese vs nonobese leukemia mouse models, leading to decreased overall drug exposure among obese mice (85). Changes in PK can have serious implications for treatment efficacy in pediatrics; for example, a retrospective study by Bhandari et al. (86) demonstrated that pediatric patients with solid tumors treated with cisplatin-containing regimens with obesity at diagnosis had a threefold greater risk for developing severe treatment-related toxicity.
The impact of body composition on PK extends beyond traditional chemotherapies. Crizotinib, a potent oral tyrosine kinase inhibitor, has increasingly been studied in pediatric solid tumors and brain tumors. In one study, overweight and/or obese status significantly influenced crizotinib PK, resulting in a clinically relevant impact (>20%) on drug exposure (87). Additionally, in a study of pediatric patients with osteosarcoma, body composition was a key determinant of exposure to bevacizumab, a humanized monoclonal antibody that inhibits vascular endothelial growth factor, and BMI was statistically significantly correlated to body-weight normalized clearance and volume of distribution, similar to studies in adult patients (88). Bevacizumab dosing adjusted for body composition in children may minimize PK variability and reduce the likelihood of major wound-healing complications. Evaluating the impact of energy balance and PK of targeted therapies is needed in the pediatric population, and future studies should be conducted as these agents become increasingly used in the pediatric cancer population.
Older adults
Most cancer diagnoses occur in adults aged older than 65 years, and given changing demographics, it is estimated that approximately 70% of all cancer diagnoses will be among older adults by 2030 (89). The management of older adults is made uniquely challenging because of large variability in treatment tolerability related to comorbid conditions and aging-related impairments that are not adequately explained by chronologic age alone (90,91). Generally, dietary intake and nutritional status change in late life. Specifically, aging may be associated with loss of appetite, taste and smell alterations, poorer oral health, reduced ability to swallow, mobility constraints, and socioeconomic barriers that contribute to poor quality and quantity of dietary intake (92). These factors collectively place older adults at high risk for poor nutritional status; in fact, 8.5% of community-dwelling older adults and 28.0% of older adults in the hospital are at risk of malnutrition (93). Cross-sectional studies indicate that total energy expenditure begins to decline around ages 50-60 years, likely driven by decreased resting and physical activity energy expenditure (94). In terms of energy storage, older adults are at increased risk for and have a higher prevalence of sarcopenia and myosteatosis, which may partly explain differences in treatment tolerability (95,96). For example, in a recent large study of adults with colorectal cancer, only 26.8% of adults aged younger than 60 years were identified as sarcopenic, whereas 58.3% of those aged older than 70 years were sarcopenic (97). Older adults diagnosed with cancer and undergoing cancer treatment also have evidence of an accelerated loss of lean mass compared with noncancer populations (98). Furthermore, the aging process is accompanied by aging-related declines in physiology and organ function, such as reduced liver size and glomerular filtration rate, that impact PK through changes in absorption, metabolism, drug distribution, and excretion (3).
Although older adults represent most of the cancer population, older adults are frequently underrepresented in clinical trials, and few studies have specifically examined this vulnerable population (99). Most studies to date have focused on mixed populations of adults, and we were unable to identify any that specifically examined the impact of energy balance on PK considerations in older adults with cancer. However, a recent perspective on chemotherapy dosing considerations in older adults identified 7 studies examining age-related PK changes among older adult populations (3). In one study by Lichtman et al. (100), paclitaxel given every 3 weeks examined age-related changes in PK and demonstrated a statistically significant association between age and rise in AUC and decline in clearance. For the growing number of vulnerable older adults diagnosed with cancer, treatment decisions are complicated by many factors, and there are many age-related considerations that may impact cancer therapeutic dosing, potentially including energy balance, but this has yet to be fully elucidated.
Future Directions
Understanding the multiple facets of energy balance and how they may impact PK of chemotherapeutics is critical to optimizing care across the cancer population. Yet, the literature is sparse, with many research gaps. More research is needed to identify which patient populations may benefit from acute dietary intake and novel nutritional strategies. For example, consideration of PK in response to various fasting protocols would help clarify the safety and therapeutic benefit of this popular dietary pattern. Further understanding of the relationship between general nutritional status and PK may also help develop and refine targeted interventions to maximize treatment response. Although the physical and psychosocial benefits outweigh the potential harms of exercise during cancer treatment, understanding the impact exercise has on PK, such as drug absorption, drug distribution, and drug metabolism, as well as cancer outcomes, is warranted. In addition, more studies examining the impact of body composition (skeletal muscle, adipose tissue, and other organs) on chemotherapeutic PK that are adequately powered and with sufficient pharmacologic endpoints are necessary to confirm preliminary findings and ultimately develop body composition–informed dosing strategies. Next, studies should focus not only on adults but also on important populations frequently underrepresented in clinical trials, particularly pediatric and older adult populations, as these are unique groups warranting specific consideration. Lastly, cotranslational approaches bridging preclinical and clinical investigations are needed to better leverage the notable gaps in both research approaches.
Acknowledgements
Role of the funder: All authors attended the said workshop supported by the funder.
Any prior presentations. None.
Contributor Information
Sarah A Purcell, Department of Medicine, Division of Endocrinology, University of British Columbia, Vancouver, Canada; Irving K. Barber Faculty of Science, Department of Biology, University of British Columbia Okanagan, Kelowna, Canada.
Dieuwertje E Kok, Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, the Netherlands.
Tyler Ketterl, Division of Pediatric Hematology/Oncology and Bone Marrow Transplant, Department of Pediatrics, University of Washington, Seattle, WA, USA; Cancer and Blood Disorders Center, Seattle Children’s Hospital, Seattle, WA, USA.
Miriam B Garcia, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
Lenat Joffe, Division of Pediatric Hematology, Oncology, and Stem Cell Transplantation, Cohen Children’s Medical Center, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, New Hyde Park, NY, USA.
Justin C Brown, Pennington Biomedical Research Center, Baton Rouge, LA, USA.
Christina M Dieli-Conwright, Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
Grant R Williams, Institute for Cancer Outcomes and Survivorship, Division of Hematology/Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
Data availability
No new data were generated or analyzed in support of this research.
Author contributions
Sarah Purcell, PhD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Dieuwertje Kok, PhD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Tyler Ketterl, PhD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Miriam Garcia, MD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Lenat Joffe, MD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Justin Brown, PhD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), Christina M. Dieli-Conwright, PhD, MPH (Conceptualization; Data curation; Writing—original draft; Writing—review & editing), and Grant Williams, MD (Conceptualization; Data curation; Writing—original draft; Writing—review & editing).
Funding
This work was supported by the TREC Training Workshop (National Cancer Institute R25CA203650; PI: Melinda Irwin).
Conflicts of interest
None declared.
References
- 1. Siegel RL, Miller KD, Fuchs HE, et al. Cancer statistics, 2022. CA Cancer J Clin. 2022;72(1):7-33. [DOI] [PubMed] [Google Scholar]
- 2. Williams GR, Al-Obaidi M, Rower J, et al. Does oxaliplatin pharmacokinetics (PKs) explain associations between body composition and chemotherapy toxicity risk in older adults with gastrointestinal (GI) cancers? J Clin Oncol. 2021;39(suppl 15):3095. [Google Scholar]
- 3. Williams GR, Outlaw D, Harvey RD, Lichtman SM, Zamboni WC, Giri S.. Chemotherapy dosing in older adults with cancer: One size does NOT fit all. J Geriatr Oncol. 2023;14(1):101363. [DOI] [PubMed] [Google Scholar]
- 4. Brown JC, Cespedes Feliciano EM, Caan BJ.. The evolution of body composition in oncology-epidemiology, clinical trials, and the future of patient care: facts and numbers. J Cachexia Sarcopenia Muscle. 2018;9(7):1200-1208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Shachar SS, Williams GR, Muss HB, et al. Prognostic value of sarcopenia in adults with solid tumours: a meta-analysis and systematic review. Eur J Cancer. 2016;57:58-67. [DOI] [PubMed] [Google Scholar]
- 6. Surov A, Pech M, Gessner D, et al. Low skeletal muscle mass is a predictor of treatment related toxicity in oncologic patients. A meta-analysis. Clin Nutr. 2021;40(10):5298-5310. [DOI] [PubMed] [Google Scholar]
- 7. Baracos VE, Arribas L.. Sarcopenic obesity: hidden muscle wasting and its impact for survival and complications of cancer therapy. Ann Oncol. 2018;29(suppl 2):ii1-ii9. [DOI] [PubMed] [Google Scholar]
- 8. Aleixo GFP, Shachar SS, Nyrop KA, et al. Myosteatosis and prognosis in cancer: systematic review and meta-analysis. Crit Rev Oncol Hematol. 2020;145:102839. [DOI] [PubMed] [Google Scholar]
- 9. Deng J, Zhu X, Chen Z, et al. A review of food-drug interactions on oral drug absorption. Drugs. 2017;77(17):1833-1855. [DOI] [PubMed] [Google Scholar]
- 10. Abuhelwa AY, Williams DB, Upton RN, et al. Food, gastrointestinal pH, and models of oral drug absorption. Eur J Pharm Biopharm. 2017;112:234-248. [DOI] [PubMed] [Google Scholar]
- 11. Singh BN, Malhotra BK.. Effects of food on the clinical pharmacokinetics of anticancer agents: underlying mechanisms and implications for oral chemotherapy. Clin Pharmacokinet. 2004;43(15):1127-1156. [DOI] [PubMed] [Google Scholar]
- 12. Food and Drug Administration. Guidance for Industry: Food-Effect Bioavailability and Fed Bioequivalence Studies.2002. https://www.fda.gov/files/drugs/published/Food-Effect-Bioavailability-and-Fed-Bioequivalence-Studies.pdf#:~:text=This%20guidance%20provides%20recommendations%20to%20sponsors%20and%2For%20applicants,and%20fed%20BE%20studies%20should%20be%20performed.%202. Accessed July 1, 2022.
- 13. Segal EM, Flood MR, Mancini RS, et al. Oral chemotherapy food and drug interactions: a comprehensive review of the literature. J Oncol Pract. 2014;10(4):e255-e268. [DOI] [PubMed] [Google Scholar]
- 14. European Medicines Agency. Guideline on the Investigation of Drug Interactions. London, UK: European Medicines Agency; 2012. [Google Scholar]
- 15. Koziolek M, Alcaro S, Augustijns P, et al. The mechanisms of pharmacokinetic food-drug interactions - a perspective from the UNGAP group. Eur J Pharm Sci. 2019;134:31-59. [DOI] [PubMed] [Google Scholar]
- 16. van Leeuwen RW, Peric R, Hussaarts KG, et al. Influence of the acidic beverage cola on the absorption of erlotinib in patients with non-small-cell lung cancer. J Clin Oncol. 2016;34(12):1309-1314. [DOI] [PubMed] [Google Scholar]
- 17. Szmulewitz RZ, Peer CJ, Ibraheem A, et al. Prospective international randomized phase II study of low-dose abiraterone with food versus standard dose abiraterone in castration-resistant prostate cancer. J Clin Oncol. 2018;36(14):1389-1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Jamei M, Turner D, Yang J, et al. Population-based mechanistic prediction of oral drug absorption. AAPS J. 2009;11(2):225-237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Muluneh B, Deal A, Alexander MD, et al. Patient perspectives on the barriers associated with medication adherence to oral chemotherapy. J Oncol Pharm Pract. 2018;24(2):98-109. [DOI] [PubMed] [Google Scholar]
- 20. Ford KL, Orsso CE, Kiss N, et al. Dietary choices following a cancer diagnosis: a narrative review. Nutrition. 2022;103-104:111838. doi: 10.1016/j.nut.2022.111838:111838. [DOI] [PubMed] [Google Scholar]
- 21. Zeeneldin AA, Taha FM.. Fasting among Muslim cancer patients during the holy month of Ramadan. Ann Saudi Med. 2012;32(3):243-249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Tas F, Karabulut S, Ciftci R, et al. The behavior of Turkish cancer patients in fasting during the holy month of Ramadan. Jpn J Clin Oncol. 2014;44(8):705-710. [DOI] [PubMed] [Google Scholar]
- 23. Heo G, Kim SH, Chang MJ.. Effect of ketogenic diet and other dietary therapies on anti-epileptic drug concentrations in patients with epilepsy. J Clin Pharm Ther. 2017;42(6):758-764. [DOI] [PubMed] [Google Scholar]
- 24. Kverneland M, Taubøll E, Molteberg E, et al. Pharmacokinetic interaction between modified Atkins diet and antiepileptic drugs in adults with drug-resistant epilepsy. Epilepsia. 2019;60(11):2235-2244. [DOI] [PubMed] [Google Scholar]
- 25. Achterbergh R, Lammers LA, van Nierop S, et al. A short-term high fat diet increases exposure to midazolam and omeprazole in healthy subjects. Expert Opin Drug Metab Toxicol. 2016;12(7):715-720. [DOI] [PubMed] [Google Scholar]
- 26. Fagan TC, Walle T, Oexmann MJ, et al. Increased clearance of propranolol and theophylline by high-protein compared with high-carbohydrate diet. Clin Pharmacol Ther. 1987;41(4):402-406. [DOI] [PubMed] [Google Scholar]
- 27. Pérez-Pitarch A, Guglieri-López B, Nacher A, et al. Impact of undernutrition on the pharmacokinetics and pharmacodynamics of anticancer drugs: a literature review. Nutr Cancer. 2017;69(4):555-563. [DOI] [PubMed] [Google Scholar]
- 28. Charland SL, Bartlett D, Torosian MH.. Effect of protein-calorie malnutrition on methotrexate pharmacokinetics. JPEN J Parenter Enteral Nutr. 1994;18(1):45-49. [DOI] [PubMed] [Google Scholar]
- 29. El-Demerdash E, Ali AA, El-Taher DE, et al. Effect of low-protein diet on anthracycline pharmacokinetics and cardiotoxicity. J Pharm Pharmacol. 2012;64(3):344-352. [DOI] [PubMed] [Google Scholar]
- 30. O’Flanagan CH, Smith LA, McDonell SB, et al. When less may be more: calorie restriction and response to cancer therapy. BMC Med. 2017;15(1):106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. van den Boogaard WMC, van den Heuvel-Eibrink MM, Hoeijmakers JHJ, et al. Nutritional preconditioning in cancer treatment in relation to DNA damage and aging. Annu Rev Cancer Biol. 2021;5:161-179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Costa EHS, Krüger JF, Camargo CQ, et al. Effects of fasting on chemotherapy treatment response: a systematic review of current evidence and suggestions for the design of future clinical trials. Nutr Cancer. 2022;74(4):1213-1221. [DOI] [PubMed] [Google Scholar]
- 33. Man FM, Eerden RAG, Doorn GM, et al. Effects of protein and calorie restriction on the metabolism and toxicity profile of irinotecan in cancer patients. Clin Pharmacol Ther. 2021;109(5):1304-1313. [DOI] [PubMed] [Google Scholar]
- 34. de Groot S, Pijl H, van der Hoeven JJM, et al. Effects of short-term fasting on cancer treatment. J Exp Clin Cancer Res. 2019;38(1):209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Printezi MI, Kilgallen AB, Bond MJG, et al. Toxicity and efficacy of chronomodulated chemotherapy: a systematic review. Lancet Oncol. 2022;23(3):e129-e143. [DOI] [PubMed] [Google Scholar]
- 36. Lau SKM, Iyengar P.. Implications of weight loss for cancer patients receiving radiotherapy. Curr Opin Support Palliat Care. 2017;11(4):261-265. [DOI] [PubMed] [Google Scholar]
- 37. Martin L, Senesse P, Gioulbasanis I, et al. Diagnostic criteria for the classification of cancer-associated weight loss. J Clin Oncol. 2015;33(1):90-99. [DOI] [PubMed] [Google Scholar]
- 38. Ottery FD. Definition of standardized nutritional assessment and interventional pathways in oncology. Nutrition. 1996;12(1):S15-S19. [DOI] [PubMed] [Google Scholar]
- 39. Williams GR, Al-Obaidi M, Dai C, et al. Association of malnutrition with geriatric assessment impairments and health-related quality of life among older adults with gastrointestinal malignancies. Cancer. 2020;126(23):5147-5155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Jager-Wittenaar H, Ottery FD.. Assessing nutritional status in cancer: role of the Patient-Generated Subjective Global Assessment. Curr Opin Clin Nutr Metab Care. 2017;20(5):322-329. [DOI] [PubMed] [Google Scholar]
- 41. Jain R, Handorf E, Khare V, et al. Impact of baseline nutrition and exercise status on toxicity and outcomes in phase I and II oncology clinical trial participants. Oncologist. 2020;25(2):161-169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Klute KA, Brouwer J, Jhawer M, et al. Chemotherapy dose intensity predicted by baseline nutrition assessment in gastrointestinal malignancies: a multicentre analysis. Eur J Cancer. 2016;63:189-200. [DOI] [PubMed] [Google Scholar]
- 43. Phippen NT, Lowery WJ, Barnett JC, et al. Evaluation of the Patient-Generated Subjective Global Assessment (PG-SGA) as a predictor of febrile neutropenia in gynecologic cancer patients receiving combination chemotherapy: a pilot study. Gynecol Oncol. 2011;123(2):360-364. [DOI] [PubMed] [Google Scholar]
- 44. Williams GR, Al-Obaidi M, Rower J, et al. Does oxaliplatin pharmacokinetics (PKs) explain associations between body composition and chemotherapy toxicity risk in older adults with gastrointestinal (GI) cancers? J Clin Oncol. 2021;39(suppl 15):3095-3095. [Google Scholar]
- 45. Purcell SA, Wallengren O, Baracos VE, et al. Determinants of change in resting energy expenditure in patients with stage III/IV colorectal cancer. Clin Nutr. 2020;39(1):134-140. [DOI] [PubMed] [Google Scholar]
- 46. Purcell SA, Elliott SA, Walter PJ, et al. Total energy expenditure in patients with colorectal cancer: associations with body composition, physical activity, and energy recommendations. Am J Clin Nutr. 2019;110(2):367-376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Boudou-Rouquette P, de Moura A, Martinez-Tapia C, et al. ; for the ELCAPA Study Group. Energy expenditure profiles and the risk of early limiting toxicity in older patients with cancer: the ELCAPA-25 prospective cohort survey. Clin Nutr. 2022;41(5):1073-1082. [DOI] [PubMed] [Google Scholar]
- 48. Demark-Wahnefried W, Hars V, Conaway MR, et al. Reduced rates of metabolism and decreased physical activity in breast cancer patients receiving adjuvant chemotherapy. Am J Clin Nutr. 1997;65(5):1495-1501. [DOI] [PubMed] [Google Scholar]
- 49. Barber MD, Mcmillan DC, Preston T, et al. Metabolic response to feeding in weight-losing pancreatic cancer patients and its modulation by a fish-oil-enriched nutritional supplement. Clin Sci. 2000;98(4):389-399. [PubMed] [Google Scholar]
- 50. Weston P, King R, Goode A, et al. Diet-induced thermogenesis in patients with gastrointestinal cancer cachexia. Clin Sci (Lond)). 1989;77(2):133-138. [DOI] [PubMed] [Google Scholar]
- 51. Pitt JP, McCarthy OM, Hoeg-Jensen T, et al. Factors influencing insulin absorption around exercise in type 1 diabetes. Front Endocrinol (Lausanne). 2020;11:573275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. McAuley SA, Horsburgh JC, Ward GM, et al. Insulin pump basal adjustment for exercise in type 1 diabetes: a randomised crossover study. Diabetologia. 2016;59(8):1636-1644. [DOI] [PubMed] [Google Scholar]
- 53. Curnier D. The potential role of exercise on the bioavailability of cancer treatments. Acad J Pediatr Neonatol. 2019;7(4):555775. [Google Scholar]
- 54. Ashcraft KA, Peace RM, Betof AS, et al. Efficacy and mechanisms of aerobic exercise on cancer initiation, progression, and metastasis: a critical systematic review of in vivo preclinical data. Cancer Res. 2016;76(14):4032-4050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Eschke RK, Lampit A, Schenk A, et al. Impact of physical exercise on growth and progression of cancer in rodents-a systematic review and meta-analysis. Front Oncol. 2019;9:35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Yang L, Morielli AR, Heer E, et al. Effects of exercise on cancer treatment efficacy: a systematic review of preclinical and clinical studies. Cancer Res. 2021;81(19):4889-4895. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Ashcraft KA, Warner AB, Jones LW, et al. Exercise as adjunct therapy in cancer. Semin Radiat Oncol. 2019;29(1):16-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Lamkin DM, Garland T Jr. Translating preclinical research for exercise oncology: take it to the VO(2max). Front Oncol. 2020;10:575657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Morrell MBG, Alvarez-Florez C, Zhang A, et al. Vascular modulation through exercise improves chemotherapy efficacy in Ewing sarcoma. Pediatr Blood Cancer. 2019;66(9):e27835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Thomas VJ, Seet-Lee C, Marthick M, et al. Aerobic exercise during chemotherapy infusion for cancer treatment: a novel randomised crossover safety and feasibility trial. Support Care Cancer. 2020;28(2):625-632. [DOI] [PubMed] [Google Scholar]
- 61. Bland KA, Zadravec K, Landry T, et al. Impact of exercise on chemotherapy completion rate: a systematic review of the evidence and recommendations for future exercise oncology research. Crit Rev Oncol Hematol. 2019;136:79-85. [DOI] [PubMed] [Google Scholar]
- 62. Ligibel JA, Bohlke K, May AM, et al. Exercise, diet, and weight management during cancer treatment: ASCO guideline. J Clin Oncol. 2022;40(22):2491-2507. [DOI] [PubMed] [Google Scholar]
- 63. Bhakta N, Liu Q, Ness KK, et al. The cumulative burden of surviving childhood cancer: an initial report from the St Jude Lifetime Cohort Study (SJLIFE). Lancet. 2017;390(10112):2569-2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Huiskamp LFJ, Chargi N, Devriese LA, et al. The predictive value of low skeletal muscle mass assessed on cross-sectional imaging for anti-cancer drug toxicity: a systematic review and meta-analysis. J Clin Med. 2020;9(11):3780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Prior SJ, Ryan AS, Blumenthal JB, et al. Sarcopenia is associated with lower skeletal muscle capillarization and exercise capacity in older adults. J Gerontol A Biol Sci Med Sci. 2016;71(8):1096-1101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Williams GR, Deal AM, Muss HB, et al. Frailty and skeletal muscle in older adults with cancer. J Geriatr Oncol. 2018;9(1):68-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Williams GR, Deal AM, Muss HB, et al. Skeletal muscle measures and physical function in older adults with cancer: sarcopenia or myopenia? Oncotarget. 2017;8(20):33658-33665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Pinkel D. The use of body surface area as a criterion of drug dosage in cancer chemotherapy. Cancer Res. 1958;18(7):853-856. [PubMed] [Google Scholar]
- 69. Baker SD, Verweij J, Rowinsky EK, et al. Role of body surface area in dosing of investigational anticancer agents in adults, 1991-2001. J Natl Cancer Inst. 2002;94(24):1883-1888. [DOI] [PubMed] [Google Scholar]
- 70. Gurney H. Dose calculation of anticancer drugs: a review of the current practice and introduction of an alternative. J Clin Oncol. 1996;14(9):2590-2611. [DOI] [PubMed] [Google Scholar]
- 71. Prado CM, Maia YL, Ormsbee M, et al. Assessment of nutritional status in cancer–the relationship between body composition and pharmacokinetics. Anticancer Agents Med Chem. 2013;13(8):1197-1203. [DOI] [PubMed] [Google Scholar]
- 72. Hopkins JJ, Sawyer MB.. A review of body composition and pharmacokinetics in oncology. Expert Rev Clin Pharmacol. 2017;10(9):947-956. [DOI] [PubMed] [Google Scholar]
- 73. Prado CM, Lima IS, Baracos VE, et al. An exploratory study of body composition as a determinant of epirubicin pharmacokinetics and toxicity. Cancer Chemother Pharmacol. 2011;67(1):93-101. [DOI] [PubMed] [Google Scholar]
- 74. Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS One. 2012;7(5):e37563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Williams GR, Deal AM, Shachar SS, et al. The impact of skeletal muscle on the pharmacokinetics and toxicity of 5-fluorouracil in colorectal cancer. Cancer Chemother Pharmacol. 2018;81(2):413-417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Schmulenson E, Zimmermann N, Muller L, et al. Influence of the skeletal muscle index on pharmacokinetics and toxicity of fluorouracil. Cancer Med. 2023;12(3):2580-2589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Molenaar-Kuijsten L, Jacobs BAW, Kurk SA, et al. Worse capecitabine treatment outcome in patients with a low skeletal muscle mass is not explained by altered pharmacokinetics. Cancer Med. 2021;10(14):4781-4789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Erdmann F, Frederiksen LE, Bonaventure A, et al. Childhood cancer: survival, treatment modalities, late effects and improvements over time. Cancer Epidemiol. 2021;71(pt B):101733. [DOI] [PubMed] [Google Scholar]
- 79. Joffe L, Shen W, Shadid G, et al. Skeletal muscle and adipose tissue changes in the first phase of treatment of pediatric solid tumors. Cancer Med. 2021;10(1):15-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Joffe L, Ladas EJ.. Nutrition during childhood cancer treatment: current understanding and a path for future research. Lancet Child Adolesc Health. 2020;4(6):465-475. [DOI] [PubMed] [Google Scholar]
- 81. Browning B, Thormann K, Donaldson A, et al. Busulfan dosing in children with BMIs ≥ 85% undergoing HSCT: a new optimal strategy. Biol Blood Marrow Transplant. 2011;17(9):1383-1388. [DOI] [PubMed] [Google Scholar]
- 82. Zuccaro P, Guandalini S, Pacifici R, et al. Fat body mass and pharmacokinetics of oral 6-mercaptopurine in children with acute lymphoblastic leukemia. Ther Drug Monit. 1991;13(1):37-41. [DOI] [PubMed] [Google Scholar]
- 83. Thompson PA, Rosner GL, Matthay KK, et al. Impact of body composition on pharmacokinetics of doxorubicin in children: a Glaser Pediatric Research Network study. Cancer Chemother Pharmacol. 2009;64(2):243-251. [DOI] [PubMed] [Google Scholar]
- 84. Orgel E, Nabais T, Douglas C, et al. Effect of body fat on population pharmacokinetics of high-dose methotrexate in pediatric patients with acute lymphoblastic leukemia. J Clin Pharmacol. 2021;61(6):755-762. [DOI] [PubMed] [Google Scholar]
- 85. Behan JW, Avramis VI, Yun JP, et al. Diet-induced obesity alters vincristine pharmacokinetics in blood and tissues of mice. Pharmacol Res. 2010;61(5):385-390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Bhandari R, Scott E, Yeh MY, et al. Association of body mass index with toxicity and survival in pediatric patients treated with cisplatin-containing regimens. Pediatr Hematol Oncol. 2021;38(3):239-250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Gibson EG, Campagne O, Selvo NS, et al. Population pharmacokinetic analysis of crizotinib in children with progressive/recurrent high-grade and diffuse intrinsic pontine gliomas. Cancer Chemother Pharmacol. 2021;88(6):1009-1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Turner DC, Navid F, Daw NC, et al. Population pharmacokinetics of bevacizumab in children with osteosarcoma: implications for dosing. Clin Cancer Res. 2014;20(10):2783-2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Smith BD, Smith GL, Hurria A, et al. Future of cancer incidence in the United States: burdens upon an aging, changing nation. J Clin Oncol. 2009;27(17):2758-2765. [DOI] [PubMed] [Google Scholar]
- 90. Williams GR, Mackenzie A, Magnuson A, et al. Comorbidity in older adults with cancer. J Geriatr Oncol. 2016;7(4):249-257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Jolly TA, Deal AM, Nyrop KA, et al. Geriatric assessment-identified deficits in older cancer patients with normal performance status. Oncologist. 2015;20(4):379-385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Nieuwenhuizen WF, Weenen H, Rigby P, et al. Older adults and patients in need of nutritional support: review of current treatment options and factors influencing nutritional intake. Clin Nutr. 2010;29(2):160-169. [DOI] [PubMed] [Google Scholar]
- 93. Leij-Halfwerk S, Verwijs MH, van Houdt S, et al. Prevalence of protein-energy malnutrition risk in European older adults in community, residential and hospital settings, according to 22 malnutrition screening tools validated for use in adults ≥65 years: a systematic review and meta-analysis. Maturitas. 2019;126:80-89. [DOI] [PubMed] [Google Scholar]
- 94. Speakman JR, Westerterp KR.. Associations between energy demands, physical activity, and body composition in adult humans between 18 and 96 y of age. Am J Clin Nutr. 2010;92(4):826-834. [DOI] [PubMed] [Google Scholar]
- 95. Williams GR, Rier HN, McDonald A, et al. Sarcopenia & aging in cancer. J Geriatr Oncol. 2019;10(3):374-377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Williams GR, Dunne RF, Giri S, et al. Sarcopenia in the older adult with cancer. J Clin Oncol. 2021;39(19):2068-2078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Caan BJ, Meyerhardt JA, Kroenke CH, et al. Explaining the obesity paradox: the association between body composition and colorectal cancer survival (C-SCANS Study). Cancer Epidemiol Biomarkers Prev. 2017;26(7):1008-1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Williams GR, Chen Y, Kenzik KM, et al. Assessment of sarcopenia measures, survival, and disability in older adults before and after diagnosis with cancer. JAMA Netw Open. 2020;3(5):e204783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Rocque GB, Williams GR.. Bridging the data-free zone: decision making for older adults with cancer. J Clin Oncol. 2019;37(36):3469-3471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Lichtman SM, Hollis D, Miller AA, et al. Prospective evaluation of the relationship of patient age and paclitaxel clinical pharmacology: Cancer and Leukemia Group B (CALGB 9762). J Clin Oncol. 2006;24(12):1846-1851. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No new data were generated or analyzed in support of this research.


