Skip to main content
Free access
ORIGINAL REPORTS
August 01, 2011

Predicting Chemotherapy Toxicity in Older Adults With Cancer: A Prospective Multicenter Study

Publication: Journal of Clinical Oncology
PDF

Abstract

Purpose

Older adults are vulnerable to chemotherapy toxicity; however, there are limited data to identify those at risk. The goals of this study are to identify risk factors for chemotherapy toxicity in older adults and develop a risk stratification schema for chemotherapy toxicity.

Patients and Methods

Patients age ≥ 65 years with cancer from seven institutions completed a prechemotherapy assessment that captured sociodemographics, tumor/treatment variables, laboratory test results, and geriatric assessment variables (function, comorbidity, cognition, psychological state, social activity/support, and nutritional status). Patients were followed through the chemotherapy course to capture grade 3 (severe), grade 4 (life-threatening or disabling), and grade 5 (death) as defined by the National Cancer Institute Common Terminology Criteria for Adverse Events.

Results

In total, 500 patients with a mean age of 73 years (range, 65 to 91 years) with stage I to IV lung (29%), GI (27%), gynecologic (17%), breast (11%), genitourinary (10%), or other (6%) cancer joined this prospective study. Grade 3 to 5 toxicity occurred in 53% of the patients (39% grade 3, 12% grade 4, 2% grade 5). A predictive model for grade 3 to 5 toxicity was developed that consisted of geriatric assessment variables, laboratory test values, and patient, tumor, and treatment characteristics. A scoring system in which the median risk score was 7 (range, 0 to 19) and risk stratification schema (risk score: percent incidence of grade 3 to 5 toxicity) identified older adults at low (0 to 5 points; 30%), intermediate (6 to 9 points; 52%), or high risk (10 to 19 points; 83%) of chemotherapy toxicity (P < .001).

Conclusion

A risk stratification schema can establish the risk of chemotherapy toxicity in older adults. Geriatric assessment variables independently predicted the risk of toxicity.

Introduction

Cancer is a disease associated with aging. Patients age 65 years and older have an 11-fold increase in cancer incidence and a 16-fold increase in cancer mortality when compared with those younger than 65 years of age.1 This population of older adults is growing rapidly. By 2030, 20% of the population in the United States will be older than age 65 years. Oncologists are ill prepared for this demographic shift, because older adults have been underrepresented in oncology clinical trials that set the standard of care.2,3 The available data suggest that older adults derive benefit from chemotherapy similar to that derived by younger adults,4,5 older age is a risk factor for chemotherapy toxicity,6 and older adults are less likely to be offered chemotherapy because of concerns regarding their ability to tolerate the treatment.7,8 Although tools have been developed to quantify chemotherapy benefit by age,9 there are no tools to characterize the risks of chemotherapy in older adults.
Currently, there is no consensus within the geriatric or oncology communities regarding a standard assessment that can identify those older adults at risk for chemotherapy toxicity. Existing oncology performance status measures (such as Karnofsky performance status [KPS]10 or Eastern Cooperative Oncology Group performance status11) are applied to all adult patients with cancer regardless of age to estimate functional status, assess eligibility for clinical trials, and predict treatment toxicity and survival.1214 However, these tools were validated in younger patients and do not address the heterogeneity in the aging process. Geriatricians perform a geriatric assessment that measures independent clinical predictors of morbidity and mortality in older adults15; however, this assessment has not typically been used in daily oncology practice to assist in decision making.
A predictive model that incorporates geriatric and oncologic correlates of vulnerability to chemotherapy toxicity in older adults could help both the healthcare provider and the patient weigh the benefits and risks of chemotherapy treatment and could serve as a platform to test interventions to decrease the risk of chemotherapy toxicity. The primary objective of this prospective longitudinal study was to develop a predictive model for grade 3 to 5 toxicity in a cohort of older adults with cancer that uses age, sociodemographic factors, tumor and treatment characteristics, laboratory data, and geriatric assessment variables. Furthermore, we assessed the predictive capability of this model for chemotherapy toxicity in comparison to KPS, a commonly used oncology performance status measure.

Patients and Methods

The Cancer and Aging Research Group Study “Determining the Utility of an Assessment Tool for Older Adults with Cancer” was open at seven participating institutions. Between November 2006 and November 2009, 500 patients were recruited from the outpatient oncology practices. Eligible patients were age ≥ 65 years, had a diagnosis of cancer, were scheduled to receive a new chemotherapy regimen, and were fluent in English (since all measures in the geriatric assessment tool were not validated in other languages). Assuming a prevalence rate of 30% for grade 3 to 5 toxicity, 500 patients would provide 80% power to detect a prevalence difference of 11% for a dichotomous predictor in logistic regression. The study was approved by the institutional review board at each participating institution. Participating patients completed the informed consent process.

Study Schema

Before initiation of the chemotherapy regimen, a geriatric assessment tool was completed. The measures in the tool are outlined in a prior publication describing the development of the tool.16 The geriatric assessment tool (Table 1) had a health care provider and a patient portion. The health care provider portion consisted of three items: the patient's KPS,10 the Timed Up & Go measure (a performance-based measure of functional status),22 and the Blessed Orientation-Memory-Concentration test23 (a screening measure of cognitive function). The patient portion consisted of self-reported measures of functional status, comorbidity, medications, nutrition, psychological state, and social support/function. A member of the health care team assisted those who needed help with completing the questionnaires.
Table 1. Domains and Measures in the Geriatric Assessment Questionnaire16
Domain Measure No. of Items Description Range of Scores Mean SD Median Range
Functional status Activities of Daily Living (subscale of MOS Physical Health)19 10 Measures limitations in a wide range of physical functions (from bathing/dressing to vigorous activities such as running) 0-100 (higher score: better physical function) 68.5 26 75.0 0-100
  Instrumental Activities of Daily Living (subscale of the OARS)20 7 Measures ability to complete activities required to maintain independence in the community (shopping, meal preparation, making telephone calls, money management) 0-14 (higher score: less need for assistance) 12.9 1.8 14 4-14
  Karnofsky Self-Reported Performance Rating Scale10 1 Global indicator of patient function determined by patient self-report ranging from “normal” to “severely disabled” 40-100 (higher score: better physical function) 85.6 13.7 90 40-100
  Karnofsky Physician-Rated Performance Rating Scale21 1 Global indicator of patient function determined by physician report ranging from “normal” to “dead” 0-100 (higher score: better physical function) 84.7 11.4 90 50-100
  No. of falls in last 6 months 1 Indicates number of times fallen in the last 6 months   0.3 0.8 0 0-6
Comorbidity Physical Health Section (subscale of the OARS)20   Presence/absence of 13 comorbid illnesses: Number of comorbid illnesses   2.5 1.7 2 0-9
Psychological state Hospital Anxiety and Depression Scale20a 14 Assesses the level of depression and anxiety experienced in the past week 0-100 (higher score: poorer psychological state) 8.3 6.0 7 0-35
Social activity MOS Social Activity Survey19 4 Measures the degree in which physical or emotional problems interfere with level of social activity 0-100 (higher score: better social activity) 56.2 22.8 58.3 0-100
Social support MOS Social Support Survey: Emotional/Information and Tangible Subscales21a 12 Measures the perceived availability of social support 0-100 (higher score: better social support) 84.9 21.3 95.8 0-100
Nutrition Body mass index 1 Weight in kg/(height in m)2   26.2 4.7 25.0 14.9-52.2
  Percent unintentional weight loss in last 6 months 1 (Unintentional weight lost in last 6 months/baseline body weight) × 100   4.7 6.2 2.0 0-32.3
Abbreviations: MOS, Medical Outcomes Study; OARS, Older American Resources and Services; SD, standard deviation.
Tumor characteristics (tumor type and stage) and pretreatment laboratory data (WBC count, hemoglobin, blood urea nitrogen, creatinine, albumin, and liver function tests) were recorded. The following treatment characteristics were captured: chemotherapy regimen, line of chemotherapy (first line or greater), the use of WBC or RBC growth factors, and the timing of initiation of WBC growth factors (primary or secondary prophylaxis). The chemotherapy dosing for the first cycle of treatment was categorized as standard or dose reduced per the National Comprehensive Cancer Network guidelines.24
The patient was followed from beginning until the end of the chemotherapy course. Toxicities were captured at each clinical encounter (scheduled or emergency visits). Two physicians (the national study principal investigator and site principal investigator) reviewed the patient's chemotherapy course to capture grade 3 to 5 chemotherapy-related toxicities (grade 3, severe; grade 4, life-threatening; and grade 5, fatal) by using the National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE), version 3.0.25 Blood values were captured as grade 3 to 5 toxicity if they met the criteria on the date of scheduled chemotherapy or at the time the patient was seeking attention because of chemotherapy toxicities.

Statistical Analyses

Descriptive analyses were performed to summarize patient, tumor, and treatment characteristics and geriatric assessment results. The incidence of the specific categories (hematologic and nonhematologic) and types of NCI CTCAE grade 3, 4, or 5 toxicity were calculated.

Model development.

A predictive model for chemotherapy toxicity was developed. The χ2 test was used to examine the association between grade 3 to 5 toxicity and the following variables: sociodemographic factors (age, sex, education, marital status, household composition, employment status, race/ethnicity), study site, cancer type (breast, GI, genitourinary [GU], gynecologic [GYN], lung, and other), cancer stage, chemotherapy dosing (standard or dose reduced), number of chemotherapy drugs (mono- or polychemotherapy), line of treatment (first line or greater), chemotherapy duration, receipt of primary prophylaxis with WBC growth factor, prechemotherapy laboratory values (WBC, hemoglobin, liver function tests, albumin, creatinine clearance [calculated by the Cockgroft and Gault,26 Jeliffe,27 Modification of Diet in Renal Disease,28 and Wright29 formulas]), and responses to the items in the Geriatric Assessment measures (Table 1).
For numerical variables, the Youden Index30 was used to identify the cut point with the highest sensitivity and specificity in classifying the presence or absence of toxicity. The variables that reached a P value of less than .1 and clinically relevant variables (chemotherapy dosing [standard or dose reduced], number of drugs [mono- or polychemotherapy], chemotherapy duration, and receipt of primary prophylaxis with WBC growth factor) were further examined in a multivariate logistic regression model by using the best subset method31 to identify the best combined sets of risk factors that best predicted chemotherapy toxicity. We evaluated the discrimination of those models by calculating the area under the receiver operation characteristic (ROC) curve and goodness-of-fit by using the Hosmer-Lemeshow test.32 Interactions among the selected risk factors were evaluated by introducing interaction terms to the model one at a time.

Developing the scoring system.

A risk score for each risk factor was calculated by dividing the β coefficient of the variable by the lowest β coefficient in the model, rounded to the nearest whole number.33,34 The sum of the scores for each patient was calculated. The sample was divided into three risk strata (low, medium, and high risk) on the basis of approximate quartiles of risk score with the middle two quartiles combined. The difference in toxicity incidence among the strata was evaluated by using the χ2 test. The discrimination and calibration of the predictive model were assessed by using the total score as a predictor of chemotherapy toxicity.

Internal validation.

The model was internally validated by using the 10-fold cross-validation process.35,36 The study sample was randomly partitioned into 10 groups, by using nine-tenths of the cohort to obtain the β coefficient and then applying the β coefficient to examine the area under the ROC curve in the tenth group. This process was repeated 10 times to obtain the average area under the ROC curve of the model. All statistical analyses were performed by using SAS 9.1 (SAS Institute, Cary, NC).

Results

Patient, Tumor, and Treatment Characteristics

The study cohort consisted of 500 patients age ≥ 65 years with stage I to IV cancer (Table 2). The mean age of participants was 73 years (standard deviation [SD], 6.2; range, 65 to 91 years) with stage I (5%), II (12%), III (22%), and IV (61%) cancer. The most common tumor types were lung (29%), GI (27%), GYN (17%), and breast (11%). Seventy percent received polychemotherapy, 76% received standard doses of chemotherapy, 71% received first-line treatment, and 18% received primary prophylaxis with WBC growth factors.
Table 2. Patient Characteristics (N = 500)
Characteristic No. of Patients % Patients
Age, years    
    65-69 175 35
    70-74 127 25
    75-79 105 21
    80-84 73 15
    85-91 20 4
Sex    
    Female 281 56
    Male 219 44
Cancer type    
    Breast 57 11
    Lung 143 29
    GI 135 27
    GYN 87 17
    GU 50 10
    Other 28 6
Cancer stage    
    I 23 5
    II 59 12
    III 109 22
    Limited 2 0
    IV/extensive 307 61
Educational level    
    Less than high school 18 4
    High school graduate 175 35
    Associate/bachelor's degree 202 40
    Advanced degree 104 21
    Missing 1 0
Marital status    
    Married 306 61
    Widowed 113 23
    Single 16 3
    Separated, divorced 65 13
Employment status    
    Full or part-time 83 17
    Retired, homemaker, unemployed 395 79
    Disabled, medical leave 21 4
    Missing 1 0
Household composition    
    Lives alone 106 21
    Lives with spouse, partner, or child 390 78
    Missing 4 1
Race/ethnicity    
    White 426 85
    Black 42 8
    Asian 26 5
    Other 6 1
Abbreviations: GU, genitourinary; GYN, gynecologic.

Geriatric Assessment Results

The mean score on the instrumental activities of daily living scale was 12.9 (SD, 1.8; range, 4 to 14), with 43% of patients requiring assistance with those activities. The mean score on the Medical Outcomes Study (MOS) Physical Health Scale was 68.5 (SD, 26; range 0 to 100), with a score of 0 indicating completely dependent and a score of 100 indicating full physical capacity. The patients' KPS ranged from 40% to 100% with 79% of patients with a status greater than 70%. Eighteen percent reported at least one fall in the last 6 months, and 90% had at least one comorbid condition. The most common comorbid conditions were hypertension (52%), arthritis (46%), heart disease (20%), and stomach disorders (19%). Twelve percent had a low body mass index of less than 22, and 18% were obese (body mass index > 30). Thirty-eight percent reported unintentional weight loss of more than 5% of body weight over the past 6 months (Table 1).

Chemotherapy Toxicity

At least one grade 3 to 5 toxicity occurred in 53% of patients (39% grade 3, 12% grade 4, and 2% grade 5 [percentages reflect the worst grade of toxicity experienced]; Table 3). Grade 3 to 5 hematologic and nonhematologic toxicity occurred in 26% and 43%, respectively. The most common grade 3 to 5 hematologic toxicities were neutropenia (11%), leucopenia (10%), and anemia (10%). The most common grade 3 to 5 nonhematologic toxicities were fatigue (16%), infection (10%), and dehydration (9%). Thirty-one percent of patients required a dose reduction during therapy, 31% had a dose delay, and 23% were hospitalized during treatment.
Table 3. Treatment-Related Adverse Events
Toxicity Type Grade 3 to 5 Grade 3 Grade 4 Grade 5*
No. % No. % No. % No. %
Hematologic and nonhematologic 265 53 197 39 58 12 10 2
Hematologic 131 26 90 18 39 8 2 0
    ANC 57 11 40 8 17 3 0 0
    WBC 49 10 41 8 8 2 0 0
    Hemoglobin 48 10 45 9 3 1 0 0
    Platelets 25 5 14 3 11 2 0 0
    Infection with abnormal ANC 10 2 7 1 1 0 2 0
Nonhematologic 217 43 184 37 25 5 8 2
    Fatigue 81 16 79 16 2 0 0 0
    Infection with normal ANC 48 10 40 8 5 1 3 1
    Dehydration 43 9 41 8 2 0 0 0
    Thrombosis/embolism 22 4 17 3 4 1 1 0
    Hyponatremia 22 4 22 4 0 0 0 0
    Diarrhea 22 4 19 4 3 1 0 0
    Hypokalemia 15 3 15 3 0 0 0 0
    Dyspnea 13 3 5 1 7 1 1 0
    Syncope 13 3 13 3 0 0 0 0
    Neuropathy 13 3 13 3 0 0 0 0
    Nausea 12 2 12 2 0 0 0 0
Abbreviation: ANC, absolute neutrophil count.
*
Additional causes of grade 5 toxicities include cardiac ischemia/infarction, liver dysfunction/failure, pneumonitis/pulmonary infiltrate, and sudden death.
The percentages for grades 3 to 5 toxicity reflect the worst grade of toxicity experienced by the individual. Since patients could have both hematologic and nonhematologic toxicity, the sum of hematologic and nonhematologic toxicities is greater than the number of all types of toxicity.
One patient had grade 5 infection with normal ANC and grade 5 thrombosis/embolism. Another patient had grade 5 infection with normal ANC and grade 5 metabolic encephalopathy.

Factors Associated With Increased Risk of Chemotherapy Toxicity

The risk factors (Table 4) associated with grade 3 to 5 toxicity in univariate analysis (P < .1) and variables deemed to be of clinical importance (chemotherapy dosing [standard or dose reduced], number of chemotherapy drugs [mono- or polychemotherapy], and primary prophylaxis with WBC growth factor) were used to derive the model for chemotherapy toxicity which includes the following risk factors: age ≥ 72 years, cancer type (GI or GU), standard dosing of chemotherapy, polychemotherapy, hemoglobin (males: < 11 g/dL; females: < 10 g/dL), creatinine clearance less than 34 mL/min (Jelliffe formula27 using ideal weight), hearing impairment described as fair or worse, ≥ one fall in the last 6 months, limited in walking one block, need for assistance in taking medications, and decreased social activities because of physical or emotional health. No significant interaction among the selected variables was found. Both the model of 11 risk factors and the model of total risk score achieved good calibration (Hosmer-Lemeshow test, P = .85 and P = .25, respectively) and discrimination (both models: ROC = 0.72; Tables 5 and 6).
Table 4. Association Between Patient Characteristics and Toxicity
Variable* Patients No Grade 3 to 5 Toxicity Grade 3 to 5 Toxicity P
No. % No. % No. %
Sociodemographics              
    Age, years              
        65 to < 72 230 46 128 54 102 38  
        ≥ 72 270 54 107 46 163 62 < .001
Tumor/treatment variables              
    Cancer type              
        Other 315 63 170 72 145 55  
        GI or GU 185 37 65 28 120 45 < .001
    Dose              
        Reduced 120 24 59 25 61 23  
        Standard 380 76 176 75 204 77 .5854
    No. of chemotherapy agents              
        Monochemotherapy 149 30 76 32 73 28  
        Polychemotherapy 351 70 159 68 192 72 .2422
    WBC growth factor (primary prophylaxis)              
        No 410 82 190 81 220 83  
        Yes 89 18 45 19 44 17  
        Missing 1 0 0 0 1 0 .4697
    RBC growth factor              
        No 420 84 202 86 218 82  
        Yes 80 16 33 14 47 18 .2609
Laboratory variables              
    Hemoglobin, g/dL              
        ≥ 10 (female), ≥ 11 (male) 429 86 215 91 214 81  
        < 10 (female), < 11 (male) 62 12 16 7 46 17  
        Missing 9 2 4 2 5 2 < .001
    Albumin, g/dL              
        > 3.6 303 61 153 65 150 57  
        ≤ 3.6 179 36 76 32 103 39  
        Missing 18 4 6 3 12 5 .0878
Geriatric assessment variables              
    Physician-rated KPS (%)              
        > 70 402 80 198 84 204 77  
        ≤ 70 86 17 33 14 53 20  
        Missing 12 2 4 2 8 3 .0666
    Timed Up & Go, seconds              
        ≤ 10 156 31 83 35 73 28  
        > 10 249 50 107 46 142 54  
        Missing 95 19 45 19 50 19 .0446
    Falls in past 6 months              
        0 407 81 204 87 203 77  
        ≥ 1 91 18 30 13 61 23  
        Missing 2 0 1 0 1 0 .003
    BMI              
        > 26.5 197 39 106 45 91 34  
        ≤ 26.5 301 60 128 54 173 65  
        Missing 2 0 1 0 1 0 .0136
    Unintentional weight loss, %              
        ≤ 6 328 66 164 70 164 62  
        > 6 170 34 71 30 99 37  
        Missing 2 0 0 0 2 0 .0809
    Chronic liver or kidney disease              
        No 476 95 227 97 249 94  
        Yes 22 4 6 3 16 6  
        Missing 2 0 2 1 0 0 .0606
    Hearing              
        Excellent/good 370 74 183 78 187 71  
        Fair/poor/deaf 123 25 47 20 76 29  
        Missing 7 1 5 2 2 1 .0303
    Mobility              
        No assistance 411 82 204 87 207 78  
        Requires assistance 89 18 31 13 58 22 .0112
    Housework              
        No assistance 320 64 162 69 158 60  
        Requires assistance 178 36 72 31 106 40  
        Missing 2 0 1 0 1 0 .0292
    Medication intake              
        No assistance 461 92 224 95 237 89  
        Requires assistance 39 8 11 5 28 11 .0143
    Vigorous activities              
        Not limited at all 64 13 39 17 25 9  
        Limited 433 87 195 83 238 90  
        Missing 3 1 1 0 2 1 .0174
    Moderate activities              
        Not limited at all 253 51 135 57 118 45  
        Limited 244 49 100 43 144 54  
        Missing 3 1 0 0 3 1 .0057
    Limited in walking several blocks              
        Not limited at all 286 57 146 62 140 53  
        Limited 208 42 85 36 123 46  
        Missing 6 1 4 2 2 1 .0251
    Limited in walking one block              
        Not limited at all 386 77 192 82 194 73  
        Limited 109 22 40 17 69 26  
        Missing 5 1 3 1 2 1 .016
    Limited in bathing and dressing              
        Not limited at all 449 90 219 93 230 87  
        Limited 49 10 15 6 34 13  
        Missing 2 0 1 0 1 0 .0156
    Decreased social activity because of health/emotional problems              
        A little, or none of the time 278 56 142 60 136 51  
        Some, most, all of the time 218 44 92 39 126 48  
        Missing 4 1 1 0 3 1 .0493
    Limited social activity compared with others your age              
        About the same, somewhat, or much less limited than others 334 67 167 71 167 63  
        Somewhat, or much more limited than others 160 32 66 28 94 35  
        Missing 6 1 2 1 4 2 .0683
Abbreviations: BMI, body mass index; GU, genitourinary; KPS, Karnofsky performance status.
*
For numerical variables, the Youden Index30 was examined to determine the cut point in the responses that had the highest sensitivity and specificity in classifying the presence or absence of toxicity. The variables that reached a P value of < .1 as well as clinically relevant variables (chemotherapy dosing [standard or dose reduced], number of chemotherapy drugs in the regimen [mono- or polychemotherapy], and receipt of primary prophylaxis with WBC growth factor) were further examined in a multivariate logistic regression model.
χ2 test was conducted for observations without missing values.
Table 5. Predictive Model
Risk Factor Prevalence Grades 3 to 5 Toxicity OR 95% CI Score
No. % No. %
Age ≥ 72 years 270 54 163 60 1.85 1.22 to 2.82 2
Cancer type GI or GU 185 37 120 65 2.13 1.39 to 3.24 2
Chemotherapy dosing, standard dose 380 76 204 54 2.13 1.29 to 3.52 2
No. of chemotherapy drugs, polychemotherapy 351 70 192 55 1.69 1.08 to 2.65 2
Hemoglobin < 11 g/dL (male), < 10 g/dL (female) 62 12 46 74 2.31 1.15 to 4.64 3
Creatinine clearance (Jelliffe, ideal weight) < 34 mL/min 44 9 34 77 2.46 1.11 to 5.44 3
Hearing, fair or worse 123 25 76 62 1.67 1.04 to 2.69 2
No. of falls in last 6 months, 1 or more 91 18 61 67 2.47 1.43 to 4.27 3
IADL: Taking medications, with some help/unable 39 8 28 72 1.50 0.66 to 3.38 1
MOS: Walking 1 block, somewhat limited/limited a lot 109 22 69 63 1.71 1.02 to 2.86 2
MOS: Decreased social activity because of physical/emotional health, limited at least sometimes 218 44 126 58 1.36 0.90 to 2.06 1
Abbreviations: GU, genitourinary; IADL, instrumental activities of daily living; MOS, Medical Outcomes Study; OR, odds ratio.
Table 6. Ability of Risk Score Versus Physician-Rated KPS to Predict Chemotherapy Toxicity
Risk Strata No Toxicity Toxicity Total P ROC
No. % No. %
By total score           < .001 0.72*
    0-5 (low) 89 70 39 30 128    
    6-9 (mid) 110 48 117 52 227    
    10-19 (high) 19 17 90 83 109    
By physician-rated KPS (%)           .19 0.53*
    90-100 125 49 128 51 253    
    80 73 49 76 51 149    
    ≤ 70 33 38 53 62 86    
Abbreviations: KPS, Karnofsky performance status; ROC, receiver operating characteristic.
*
Risk score and physician-rated KPS were treated as continuous to calculate the ROC.
Risk scores were assigned to each risk factor, as described in the Statistical Analyses section (Table 5). The median overall risk score was 7 (range, 0 to 19). The cohort was divided into three categories on the basis of the risk of grade 3 to 5 toxicity: low risk (0 to 5 points, 30%), intermediate risk (6 to 9 points, 52%), and high risk (10 to 19 points, 83%). There was a significant difference in toxicity among the risk groups (P < .001; Fig 1 and Table 6).
Fig 1. Ability of (A) risk score versus (B) physician-rated Karnofsky performance status (KPS) to predict chemotherapy toxicity. Graphs show grade 3 to 5 toxicity.
Exploratory analyses were performed to calculate the ROC of the model by using the total risk score for each tumor type: GI (0.72), GU (0.76), breast (0.66), lung (0.68), GYN (0.66), and other (0.81) cancers.

Ability of the Model to Predict Toxicity in Comparison With KPS

The association between KPS and chemotherapy toxicity is described in Figure 1 and Table 6. There was no significant difference in the incidence of toxicity across the KPS-based risk groups (P = .19). The ROC of the model with KPS (as a continuous variable) was 0.53 which was lower than the ROC of the risk score model, 0.72. Furthermore, the addition of KPS to our final model did not improve the ROC.

Internal Validation of the Predictive Model

A 10-fold cross validation yielded an area under the curve statistic of 0.72, indicating that the model retained a good discrimination.

Discussion

This prospective multicenter study demonstrated that chemotherapy toxicity is common in older adults, with 53% experiencing at least one grade 3 to 5 toxicity. Among these, 2% experienced a treatment-related mortality. A predictive model was developed to identify those patients at greatest risk, including factors obtained in everyday practice (patient age, number of chemotherapy drugs, dosing, and laboratory values) and factors not typically used in everyday oncology practice (geriatric assessment variables). This model had a greater ability to discriminate risk of chemotherapy toxicity than the KPS, which is commonly used in oncology practice.
Older adults are at increased risk for chemotherapy toxicity; however, oncologists are left with little guidance when it comes to identifying risk factors other than chronologic age. It is generally recognized that chronologic age does not equate to physiologic age. Geriatricians perform a geriatric assessment to identify clinical predictors of morbidity and mortality15; however, this assessment has not been routinely incorporated into oncology care because of the time and resource requirements. Furthermore, there is a lack of guidelines regarding how to interpret the findings in the context of oncology care.
The predictive model identified patient age, tumor, treatment, laboratory values, and geriatric assessment variables as risk factors for chemotherapy toxicity. There is a rational explanation for why each of these factors may predict chemotherapy toxicity. Although older age is associated with an accumulation of physiologic deficit, there is controversy about which chronologic age defines an individual as “older.” Age ≥ 72 years as a risk factor for chemotherapy toxicity provides evidence for the seventh decade of life as a time when the cumulative effects of aging are associated with increased vulnerability.
Tumor and treatment variables were identified as risk factors for chemotherapy toxicity. Patients with GI and GU cancers were at increased risk for toxicity, possibly reflective of the type of chemotherapy delivered or alterations in physiology (diarrhea/impaired fluid balance) associated with the cancer or the treatment. Receipt of polychemotherapy and/or standard dosing of chemotherapy were associated with an increased risk of toxicity. Aging is associated with decreased bone marrow reserve and an increased risk of myelosuppressive-associated complications from chemotherapy.37,38 The receipt of polychemotherapy further increases the risk of myelosuppressive effects from chemotherapy and can potentially amplify the physiologic stress of a regimen secondary to overlapping toxicities.
Laboratory values (anemia and renal dysfunction) were identified as risk factors for chemotherapy toxicity. The presence of anemia can further increase susceptibility to myelosuppression with certain antineoplastic drugs that are heavily bound to RBCs (epipodophyllotoxins, anthracyclines, camptothecins) by increasing the volume of distribution of these drugs.39 In the geriatric population, anemia is an independent predictor of hospitalization and mortality, perhaps representing a global measure of decreased reserve.40 There is an age-related decrease in renal function which could impact the pharmacokinetics of renally metabolized drugs.17
Geriatric assessment variables were a critical part of the predictive model. Among geriatric patients, functional status is a strong predictor of morbidity and mortality.18 Four questions that reflected the patient's functional status were included in the model (ability to walk one block, decreased social activities because of physical or emotional problems, falls in the last 6 months, and the need for assistance with taking medications). The need for assistance with taking medications could also be a surrogate measure of cognitive function, grip strength (unable to open the bottle), or vision (unable to see the instructions). A decrease in social activities because of physical or emotional problems may represent both a functional measure and a measure of psychological state. Finally, poor hearing was identified as a risk factor for chemotherapy toxicity, potentially reflecting whether the patient could hear the instructions regarding potentialadverse effects, supportive care medications, and indications of when to seek medical attention.
These findings contribute to an ongoing paradigm shift in oncology assessment. The commonly used oncology performance status measure (KPS) did not identify older adults at increased risk for chemotherapy toxicity, reflecting the limitations of trying to use one global assessment measure of functional status to describe the heterogeneity in the geriatric population. Furthermore, the KPS might be misleading. In older adults it is difficult to discriminate between a KPS of 80% (“normal activity with effort; some signs or symptoms of disease”) and a KPS of 60% (“requires occasional assistance, but is able to care for most of his/her needs”).
There are limitations to this study. This study reported on grade 3 to 5 toxicity; however, some grade 2 toxicities (diarrhea, neuropathy) may also be relevant to the geriatric population. Our study population was heterogeneous, consisting of patients with different tumor types and treatment regimens. Our rationale behind studying a heterogeneous population was to determine whether there are common factors that are predictive of vulnerability in the geriatric oncology population; however, there may be additional or different risk factors that are predictive of toxicity based on tumor type or treatment regimen. Exploratory analyses revealed that the ROC of the model was similar when applied to the different tumor types; however, our future research will focus on refining the model among patients with specific tumor types who are receiving specific treatment regimens. Finally, although the model was internally validated, these findings need to be validated externally in an independent cohort.
This study fills critical gaps in the knowledge of predictors for chemotherapy toxicity in older adults, something that does not currently exist and for which there is an enormous and growing need. It unites the fields of geriatrics and oncology by incorporating geriatric correlates of vulnerability, studying their impact in a diverse population of older adults with cancer, and identifying common risk factors for chemotherapy toxicity. Ultimately, these data will provide the basis for future intervention studies aimed at decreasing the risk of chemotherapy toxicity and maintaining the function and health of older adults with cancer.
Written on behalf of the Cancer and Aging Research Group.

Authors' Disclosures of Potential Conflicts of Interest

Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.
Employment or Leadership Position: None Consultant or Advisory Role: Arti Hurria, Amgen (C), Genentech (C), GTx (C) Stock Ownership: None Honoraria: None Research Funding: Arti Hurria, Abraxis BioScience (Celgene), GlaxoSmithKline Expert Testimony: None Other Remuneration: None

References

1.
BD Smith, GL Smith, A Hurria, etal : Future of cancer incidence in the United States: Burdens upon an aging, changing nation J Clin Oncol 27: 2758– 2765,2009
2.
LF Hutchins, JM Unger, JJ Crowley, etal : Underrepresentation of patients 65 years of age or older in cancer-treatment trials N Engl J Med 341: 2061– 2067,1999
3.
L Talarico, G Chen, R Pazdur: Enrollment of elderly patients in clinical trials for cancer drug registration: A 7-year experience by the US Food and Drug Administration J Clin Oncol 22: 4626– 4631,2004
4.
HB Muss, S Woolf, D Berry, etal : Adjuvant chemotherapy in older and younger women with lymph node-positive breast cancer JAMA 293: 1073– 1081,2005
5.
DJ Sargent, RM Goldberg, SD Jacobson, etal : A pooled analysis of adjuvant chemotherapy for resected colon cancer in elderly patients N Engl J Med 345: 1091– 1097,2001
6.
HB Muss, DA Berry, C Cirrincione, etal : Toxicity of older and younger patients treated with adjuvant chemotherapy for node-positive breast cancer: The Cancer and Leukemia Group B Experience J Clin Oncol 25: 3699– 3704,2007
7.
A Hurria, FL Wong, D Villaluna, etal : Role of age and health in treatment recommendations for older adults with breast cancer: The perspective of oncologists and primary care providers J Clin Oncol 26: 5386– 5392,2008
8.
AB Kornblith, M Kemeny, BL Peterson, etal : Survey of oncologists' perceptions of barriers to accrual of older patients with breast carcinoma to clinical trials Cancer 95: 989– 996,2002
9.
PM Ravdin, LA Siminoff, GJ Davis, etal : Computer program to assist in making decisions about adjuvant therapy for women with early breast cancer J Clin Oncol 19: 980– 991,2001
10.
DA Karnofsky, JH Burchenal: CM Macleod The clinical evaluation of chemotherapeutic agents in cancer Evaluation of Chemotherapeutic Agents 191– 205,1948 New York, NY Columbia University Press
11.
C Zubrod, M Schneiderman, E Frei III, etal : Appraisal of methods for the study of chemotherapy of cancer in man: Comparative therapeutic trial of nitrogen mustard and triethylene thiophosphoramide J Chron Dis 11: 7– 33,1960
12.
DF Bajorin, PM Dodd, M Mazumdar, etal : Long-term survival in metastatic transitional-cell carcinoma and prognostic factors predicting outcome of therapy J Clin Oncol 17: 3173– 3181,1999
13.
RJ Motzer, J Bacik, LH Schwartz, etal : Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma J Clin Oncol 22: 454– 463,2004
14.
KS Albain, JJ Crowley, M LeBlanc, etal : Survival determinants in extensive-stage non-small-cell lung cancer: The Southwest Oncology Group experience J Clin Oncol 9: 1618– 1626,1991
15.
M Extermann, A Hurria: Comprehensive geriatric assessment for older patients with cancer J Clin Oncol 25: 1824– 1831,2007
16.
A Hurria, S Gupta, M Zauderer, etal : Developing a cancer-specific geriatric assessment: A feasibility study Cancer 104: 1998– 2005,2005
17.
RD Lindeman, J Tobin, NW Shock: Longitudinal studies on the rate of decline in renal function with age J Am Geriatr Soc 33: 278– 285,1985
18.
DB Reuben, LV Rubenstein, SH Hirsch, etal : Value of functional status as a predictor of mortality: Results of a prospective study Am J Med 93: 663– 669,1992
19.
AL Stewart, JE Ware Jr: Measuring Functioning and Well-Being: The Medical Outcomes Study Approach 1992 Durham, NC Duke University Press
20.
GG Fillenbaum, MA Smyer: The development, validity, and reliability of the OARS multidimensional functional assessment questionnaire J Gerontol 36: 428– 434,1981
20a.
AS Zigmond, RP Snaith: The hospital anxiety and depression scale Acta Psychiatr Scand 67: 361– 370,1983
21.
CL Loprinzi, JA Laurie, HS Wieand, etal : Prospective evaluation of prognostic variables from patient-completed questionnaires: North Central Cancer Treatment Group J Clin Oncol 12: 601– 607,1994
21a.
CD Sherbourne, AL Stewart: The MOS social support survey Soc Sci Med 32: 705– 714,1991
22.
D Podsiadlo, S Richardson: The timed “Up & Go”: A test of basic functional mobility for frail elderly persons J Am Geriatr Soc 39: 142– 148,1991
23.
C Kawas, H Karagiozis, L Resau, etal : Reliability of the Blessed Telephone Information-Memory-Concentration Test J Geriatr Psychiatry Neurol 8: 238– 242,1995
24.
NCCN Practice Guidelines in Oncology, 2010 National Comprehensive Cancer Network http://www.nccn.org/professionals/physician_gls/f_guidelines.asp
25.
National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE), Version 3.0 http://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcaev3.pdf
26.
DW Cockcroft, MH Gault: Prediction of creatinine clearance from serum creatinine Nephron 16: 31– 41,1976
27.
RW Jelliffe: Estimation of creatinine clearance when urine cannot be collected Lancet 1: 975– 976,1971
28.
AS Levey, JP Bosch, JB Lewis, etal : A more accurate method to estimate glomerular filtration rate from serum creatinine: A new prediction equation— Modification of Diet in Renal Disease Study Group Ann Intern Med 130: 461– 470,1999
29.
JG Wright, AV Boddy, M Highley, etal : Estimation of glomerular filtration rate in cancer patients Br J Cancer 84: 452– 459,2001
30.
WJ Youden: Index for rating diagnostic tests Cancer 3: 32– 35,1950
31.
RR Hocking, RN Leslie: Selection of the best subset in regression analysis Technometrics 9: 531– 540,1967
32.
DW Hosmer, S Lemeshow: Applied Logistic Regression 2000 New York, NY Wiley
33.
J Concato, AR Feinstein, TR Holford: The risk of determining risk with multivariable models Ann Intern Med 118: 201– 210,1993
34.
LC Walter, RJ Brand, SR Counsell, etal : Development and validation of a prognostic index for 1-year mortality in older adults after hospitalization JAMA 285: 2987– 2994,2001
35.
T Hastie, R Tibshirani, J Friedman: The Elements of Statistical Learning: Data Mining, Inference, and Prediction (ed 2) 2009 New York, NY Springer
36.
DL Olsen, D Denlen: Advanced Data Mining Techniques 2008 New York, NY Springer
37.
EC Dees, S O'Reilly, SN Goodman, etal : A prospective pharmacologic evaluation of age-related toxicity of adjuvant chemotherapy in women with breast cancer Cancer Invest 18: 521– 529,2000
38.
H Gómez, M Hidalgo, L Casanova, etal : Risk factors for treatment-related death in elderly patients with aggressive non-Hodgkin's lymphoma: Results of a multivariate analysis J Clin Oncol 16: 2065– 2069,1998
39.
L Repetto, I Carreca, D Maraninchi, etal : Use of growth factors in the elderly patient with cancer: A report from the Second International Society for Geriatric Oncology (SIOG) 2001 meeting Crit Rev Oncol Hematol 45: 123– 128,2003
40.
GJ Izaks, RG Westendorp, DL Knook: The definition of anemia in older persons JAMA 281: 1714– 1717,1999

Information & Authors

Information

Published In

Journal of Clinical Oncology
Pages: 3457 - 3465
PubMed: 21810685

Presented at

Presented at the 46th Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, June 4-8, 2010.

History

Published online: August 01, 2011
Published in print: September 01, 2011

Permissions

Request permissions for this article.

Authors

Affiliations

Arti Hurria [email protected]
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Kayo Togawa
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Supriya G. Mohile
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Cynthia Owusu
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Heidi D. Klepin
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Cary P. Gross
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Stuart M. Lichtman
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Ajeet Gajra
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Smita Bhatia
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Vani Katheria
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Shira Klapper
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Kurt Hansen
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Rupal Ramani
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
Mark Lachs
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
F. Lennie Wong
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.
William P. Tew
Arti Hurria, Kayo Togawa, Smita Bhatia, Rupal Ramani, and F. Lennie Wong, City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte; Kurt Hansen, University of California Davis School of Medicine, Sacramento, CA; Supriya G. Mohile, University of Rochester Medical Center, Rochester; Stuart M. Lichtman, Shira Klapper, and William P. Tew, Memorial Sloan-Kettering Cancer Center; Mark Lachs, Weill Cornell Medical College, New York; Ajeet Gajra, State University of New York Upstate Medical University and Veterans Administration Medical Center, Syracuse, NY; Cynthia Owusu, Case Western Reserve University, Cleveland, OH; Heidi D. Klepin, Wake Forest University School of Medicine, Winston-Salem, NC; Cary P. Gross, Yale Comprehensive Cancer Center, New Haven, CT; and Vani Katheria, Georgetown University, Washington, DC.

Notes

Corresponding author: Arti Hurria, MD, City of Hope, 1500 E. Duarte Rd, Duarte, CA 91010; e-mail: [email protected].

Author Contributions

Conception and design: Arti Hurria, Mark Lachs
Financial support: Arti Hurria
Administrative support: Arti Hurria
Provision of study materials or patients: Arti Hurria, Supriya G. Mohile, Cynthia Owusu, Heidi D. Klepin, Cary P. Gross, Stuart M. Lichtman, Ajeet Gajra, William P. Tew
Collection and assembly of data: Arti Hurria, Kayo Togawa, Supriya G. Mohile, Cynthia Owusu, Heidi D. Klepin, Cary P. Gross, Stuart M. Lichtman, Ajeet Gajra, Vani Katheria, Shira Klapper, Kurt Hansen, Rupal Ramani, William P. Tew
Data analysis and interpretation: Arti Hurria, Kayo Togawa, Supriya G. Mohile, Cynthia Owusu, Heidi D. Klepin, Cary P. Gross, Stuart M. Lichtman, Ajeet Gajra, Smita Bhatia, F. Lennie Wong, William P. Tew
Manuscript writing: All authors
Final approval of manuscript: All authors

Disclosures

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

Funding Information

Supported by Paul Beeson Career Development Award in Aging Research No. K23 AG026749-01 (A.H.), Paul Beeson Career Development Award No. 1 K08 AG24842 (C.P.G.), and American Society of Clinical Oncology, Association of Specialty Professors, Junior Development Award in Geriatric Oncology (A.H.).

Metrics & Citations

Metrics

Altmetric

Citations

Article Citation

Download Citation

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Simply select your manager software from the list below and click Download.

For more information or tips please see 'Downloading to a citation manager' in the Help menu.

Format





Download article citation data for:
Arti Hurria, Kayo Togawa, Supriya G. Mohile, Cynthia Owusu, Heidi D. Klepin, Cary P. Gross, Stuart M. Lichtman, Ajeet Gajra, Smita Bhatia, Vani Katheria, Shira Klapper, Kurt Hansen, Rupal Ramani, Mark Lachs, F. Lennie Wong, William P. Tew
Journal of Clinical Oncology 2011 29:25, 3457-3465

View Options

View options

PDF

View PDF

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login

Purchase Options

Purchase this article to get full access to it.

Purchase this Article

Subscribe

Subscribe to this Journal
Renew Your Subscription
Become a Member

Media

Figures

Other

Tables

Share

Share

Share article link

Share