Citation: Alexander B. Dillon, Kevin Lin, Andrew Kwong, Susana Ortiz. Immunotherapy in Melanoma, Gastrointestinal (GI), and Pulmonary Malignancies[J]. AIMS Public Health, 2015, 2(1): 86-114. doi: 10.3934/publichealth.2015.1.86
[1] | Mohammed Adeeb Shahin, Sami Abdo Radman Al-Dubai, Duoaa Seddiq Abdoh, Abdullah Saud Alahmadi, Ahmed Khalid Ali, Tamer Hifnawy . Burnout among nurses working in the primary health care centers in Saudi Arabia, a multicenter study. AIMS Public Health, 2020, 7(4): 844-853. doi: 10.3934/publichealth.2020065 |
[2] | Van De Tran, Trung Tin Pham, Trung Hieu Le, Thanh Thao Nguyen Thi, Minh Trung Nguyen, Duong Phuc Phan, Thi Bich Thuy Bui, Minh Cuong Nguyen, Rebecca Susan Dewey, Nguyet Tu Tran . Workplace wellbeing in community pharmacy practice: A cross-sectional study in Can Tho, Vietnam. AIMS Public Health, 2024, 11(1): 258-272. doi: 10.3934/publichealth.2024013 |
[3] | Joyce Lo, Sharan Jaswal, Matthew Yeung, Vijay Kumar Chattu, Ali Bani-Fatemi, Aaron Howe, Amin Yazdani, Basem Gohar, Douglas P. Gross, Behdin Nowrouzi-Kia . A systematic review of the literature: Gender-based violence in the construction and natural resources industry. AIMS Public Health, 2024, 11(2): 654-666. doi: 10.3934/publichealth.2024033 |
[4] | Nicola Magnavita, Francesco Marcatto, Igor Meraglia, Giacomo Viti . Relationships between individual attitudes and occupational stress. A cross-sectional study. AIMS Public Health, 2025, 12(2): 557-578. doi: 10.3934/publichealth.2025030 |
[5] | Behdin Nowrouzi-Kia, Ali Bani-Fatemi, Aaron Howe, Simrat Ubhi, Mitchel Morrison, Harseerat Saini, Vijay Kumar Chattu . Examining burnout in the electrical sector in Ontario, Canada: A cross-sectional study. AIMS Public Health, 2023, 10(4): 934-951. doi: 10.3934/publichealth.2023060 |
[6] | Francesco Marcatto, Donatella Ferrante, Mateusz Paliga, Edanur Kanbur, Nicola Magnavita . Behavioral dysregulation at work: A moderated mediation analysis of sleep impairment, work-related stress, and substance use. AIMS Public Health, 2025, 12(2): 290-309. doi: 10.3934/publichealth.2025018 |
[7] | Genevieve N Healy, Ana Goode, Diane Schultz, Donna Lee, Bell Leahy, David W Dunstan, Nicholas D Gilson, Elizabeth G Eakin. . The BeUpstanding ProgramTM: Scaling up the Stand Up Australia Workplace Intervention for Translation into Practice. AIMS Public Health, 2016, 3(2): 341-347. doi: 10.3934/publichealth.2016.2.341 |
[8] | Andrew Brinkley, Josie Freeman, Hilary McDermott, Fehmidah Munir . What are the Facilitators and Obstacles to Participation in Workplace Team Sport? A Qualitative Study. AIMS Public Health, 2017, 4(1): 94-126. doi: 10.3934/publichealth.2017.1.94 |
[9] | Petros Galanis, Ioannis Moisoglou, Aglaia Katsiroumpa, Panayota Sourtzi . Impact of workplace bullying on job burnout and turnover intention among nursing staff in Greece: Evidence after the COVID-19 pandemic. AIMS Public Health, 2024, 11(2): 614-627. doi: 10.3934/publichealth.2024031 |
[10] | Carol Nash . The Health Narratives Research Group (HeNReG): A self-direction process offered to help decrease burnout in public health nurse practitioners. AIMS Public Health, 2024, 11(1): 176-208. doi: 10.3934/publichealth.2024009 |
Trust is critical to the patient-provider relationship and is considered essential for effective health care delivery [1]. Patients' trust in their physicians is associated with satisfaction with care, information disclosure of sensitive information, adherence to treatments and continuity of care [1,2]. African Americans are documented as one of the racial/ethnic minority populations with the lowest levels of trust in physicians [2,3]. African Americans' low level of trust is frequently cited to explain their persistent and disproportionate burden of adverse health and healthcare outcomes [2,4,5]. It is also well documented that discrimination and prejudice/bias on the part of health care providers has perpetuated African Americans' low trust.
Research suggest that patient-provider race concordance (i.e., when patient and provider share the same race/ethnicity) may improve African Americans' trust and consequently their health outcomes [6,7]. Patient-provider race concordance has been linked to higher trust, better communication, more shared medical decision making, higher satisfaction, greater use of medical care, and less perceived stigma and discrimination in the delivery of medical care [7,8,9,10,11,12]. African-Americans' have reported lower levels of trust with racially discordant providers [5,13] and prefer providers of the same race/ethnicity [8,12]. Nonetheless, reviews of the literature on patient-provider race concordance have concluded that strength of the evidence is modest in suggesting that ethnic minority patients would prefer and trust providers of similar racial backgrounds, or that patient-provider race-concordance is associated with more positive health outcomes [14,15,16].
Race/ethnicity is only one aspect of the human identity, which includes mutual aspects such as gender, age, and language [6]. Studies show that other types of concordance (including gender, age and language) concordance influence interpersonal care ratings of providers [6,15,17,18]. Some patients have been shown to prefer doctors of the same gender [17,18], with women more likely than men to prefer a provider of the same gender [18]. For example, a study of emergency room patients showed that women trusted female physicians more than male physicians and were more satisfied with their care [19]. The same pattern was not observed for men. Preferences and ratings have been linked to patient-physician communication and delivery of patient-centered care [20,21]. However, the studies on patient preference and interpersonal ratings of care (which includes trust and satisfaction) neither included African Americans nor examined the mutual influence of race, gender, and age concordance on patients' preference and interpersonal ratings of care for physicians.
Understanding and eliminating racial/ethnic health disparities, particularly among African Americans, are amongst the most urgent problems facing our society today. Increasing the diversity of the healthcare workforce is proposed as a solution towards decreasing racial/ethnic disparities in health and healthcare [14,15]. It is asserted that patients are more likely to choose physicians of similar racial/ethnic background when given the option [7,9,22]. But, it remains unclear whether African Americans would prefer a physician of similar racial/ethnic identity or how other aspects of the human identity (e.g., gender, age) may influence their preferences.
This study uses a factorial survey design based on social judgment theory [23] to assess older African American women's perceived trustworthiness of physicians based on manipulations of race/ethnicity, gender, and age of physicians. It also examines whether type of medical visit scenario influences perceptions of a physician's trustworthiness. Trust and concordance are complex concepts [6]. The factorial survey is specifically intended to clarify people's values and is a powerful technique for studying and analyzing people's choices and judgments about social phenomena that are complex or multidimensional in nature [24]. Older African American women constitute an expanding part of the elderly (and sickest) population in the United States [25]; hence, it is prudent to understand what influences older African Americans' trust in providers given its importance in adherence, disease management and healthy aging [26].
This study utilizes the factorial survey technique, an approach which combines experimental design and survey research methods where short descriptions of situations or persons (i.e., vignettes) are provided to participants within surveys in order to elicit their opinions or judgments about each vignette provided [23,27,28]. People routinely make judgments with consideration of various factors. The factorial survey is effective in identifying variability and patterns associated with factors used in making judgments. The factorial survey approach has been used to assess multidimensional phenomena such as preference for shared decision making in the medical encounter, preferences for racial integration, and racial/ethnic childcare preferences [29,30].
In our factorial survey, participants are presented with two vignettes of an initial physician visit: one vignette describes a routine medical checkup with no serious concerns, while the other vignette describes a visit in which the woman had felt a lump in her breast. Each vignette is accompanied with a photograph of an imaginary physician and participants are asked to judge the physician on perceived fidelity, competence, honesty, confidentiality, and global trust. The race/ethnicity, age, and gender of the imaginary physicians were randomly manipulated. This was a factorial balanced incomplete design; women were not exposed to all possible imaginary physicians. Each woman was given four imaginary physicians to evaluate. Women were given the routine visit vignette twice with two different imaginary physicians and the serious health concern visit vignette twice with two different imaginary physicians. Women had the choice of listening to or reading the vignette on the computer. Pictures of the imaginary physician and vignette were linked to a response instrument in the simulation.
Thirty-two individuals were recruited to be used in pictures as imaginary physicians. Each individual wore a white lab coat and stethoscope and was photographed against a blue background. Eight photographs were selected to use in the survey. The photograph selection was based on the highest averaged score of attractiveness, likability, and professionalism from six African American women (aged 45 and over) in the community. Seven items from the Reysen Likeability scale was used to assess, rank, and select physicians [31].
The following is the introduction script for the serious medical visit that participants received before being presented with the picture of the imaginary physician and trust question. The non-serious or routine visit does not include the statement “You are very concerned about this visit because you (think you have a lump in your breast or) think you felt a lump in your breast.”
You have been a patient at the community clinic for one year. The practice has a good reputation and you like the care that you have received. However, you do not always see the same doctor at every visit. You are scheduled to see a doctor today for an annual physical exam, which will include a breast exam. You are very concerned about this visit because you (think you have a lump in your breast or) think you felt a lump in your breast. The receptionist greets you and asks you to update any personal information in your record. You wait approximately 15 minutes; then the nurse takes you back to an examination room. The nurse reviews some information regarding your past medical history. Your blood pressure, temperature, and pulse are taken, and then you are asked to undress from your waist up and put on a gown. The next person to greet you is this doctor…
A convenience sample of 313 African American women aged 45-64 years residing in a Midwestern city was recruited. African Americans are a difficult population to recruit into epidemiologic studies [32], therefore, women were recruited actively through churches, community health centers, hair salons, health fairs, community events, senior centers and housing establishments, and through advertisements in the African American community. Women were also recruited through referral by other study participants. While these community-based recruitment methods may compromise sample representativeness, these methods have proved successful in recruiting African Americans into epidemiologic studies [32]. The sample size required for this survey was calculated on the assumption that African American women would have an average score of 3.98 on the trust measures for White physicians (with a standard deviation of 1.00), and projected that they would have 5% increase in trust when exposed to African American physicians. These sampling criteria are based on a statistical power of approximately 0.8 with an alpha = 0.05 [33].
The factorial survey was administered via laptops in university and community settings. Older and disabled women were surveyed in their homes (n = 40). Participants were also assisted with the reading of the survey and laptop technology was used when needed. Participants received a $20 gift certificate for completing the surveys. This study was approved by the University of Wisconsin-Madison Social and Behavioral Science Institutional Review Board.
Judgments about physicians were made based on five measures of trust (i.e., fidelity, competence, honesty, confidentiality, and global trust) from the Wake Forest physician trust scale [34]. Ratings range from 1 to 5 (strongly agree to strongly disagree). Trust measures are as follows: I think this doctor will do whatever it takes to get me all the care that I need (fidelity), I think this doctor will be totally honest about the care I need (honesty), I think this doctor will keep all my information private (confidentiality), I think this doctor will be extremely thorough and careful (competence), and I think I can trust this doctor to put my medical needs above all other considerations when treating my medical problems (global trust). The five items were assessed separately and as an index (i.e., summed and averaged) of trust in each doctor [35].
Profile characteristics of imaginary physicians: Women were presented with pictures which portrayed the following profile characteristics of physicians: African American young male (AAYM), African American older male (AAOM), African American young female (AAYF), African American older female (AAOF), White young male (WYM), White older male (WOM), White young female (WYF), and White older female (WOF). Young was categorized as age 30-45 years and older as 50+ years.
Respondents' characteristics: Covariates included in our analysis are based on conceptual and empirical literature indicating that trust in doctors is related to patient sociodemographic characteristics, health status, access to care, and medical care factors [1]. Covariates included as confounders were: age (45-64, 65+), education level (less than high school, high school, some college, college and above), income level (<$25,000, $25,000-$49,999, $50,000-$74,999, ≥$75,000), insurance status (insured, uninsured) perceived health status (poor/fair, good, and very good/excellent), usual source of care (yes/no), and negative experience with a health provider (yes/no).
Subjects' perceptions of trust in the 8 vignettes were analyzed using factorial survey methods [36], a technique for applying experimental design to survey research. This method is used primarily to study participants' responses to hypothetical situations, such as manipulated vignettes. MLWin software [37] was used to construct a two-level hierarchical linear model (vignettes nested within participants). This approach poses two regression models simultaneously: one modeling the vignette and scenario effects within the subjects, and one modeling the subject effect between subjects. This approach allows the partitioning of the total dispersion in the estimated regression parameters into a sampling variance and a residual variance. Through simultaneous use of information from individual respondents and the entire group of respondents, hierarchical models produce more precise parameter estimates than other methods [36]. Other analytic approaches, such as a single-level analysis and a separate-level, two-stage analysis were not used, because their assumptions are violated by factorial survey designs.
The hierarchical linear analysis was conducted on vignette variables and the severity scenario condition using multiple outcome measures of trust. Seven dummy variables (k-1 dummies) were used to represent the different vignette conditions, using the African American older female vignette as the reference group. Another dummy variable was used to represent the severity level of the scenario, with the non-serious scenario used as reference. Parameter estimates were obtained by an iterative generalized least-squares estimator. All vignette variables were included in the within-respondents portion of the model, and crossed with the severity scenario condition. All subject characteristics (age, education, income, health status, insurance coverage, usual doctor, and negative experiences encountered) were also incorporated into the subject's intercept in the between-subject portion of the model (See the Appendix in Brown, Brown, Edwards, and Nutz, 1992 for more details) [38]. Means, p-values, effect size estimations (Cohen's d) with 95% confidence intervals were used to assess effects. Results were considered statistically significant at p-value < 0.05. The GLMM models were constructed in NCSS Version 7 [39].
Characteristics of the study participants are presented in Table 1. Approximately 27% of the women were elderly (65+ years), 18.2% had less than a high school diploma, 6.4% had no usual source of care, 45.7% had an income of <$25,000, 16% had no health insurance, and 44% reported having had a negative experience with the health care system. The unadjusted overall mean scores were higher for confidentiality and honesty. Scores are based on a scale of 1 to 5, with higher scores indicating higher trust. While results from the bivariate analysis are not shown, the white-older-male had a significantly higher unadjusted mean score than the African American older female (AA-older-female) on competence (3.94, 3.71; p = 0.034) and the AA-older-male had a significantly higher unadjusted mean score than the AA-older-female on honesty (4.08, 3.86; p = 0.049). The AA-older-female was no different than counterparts on fidelity, confidentiality, global trust, or overall (i.e., index of trust) trust.
Figure 1 shows the adjusted mean (x) scores and the effect size estimations with confidence intervals from the hierarchical linear analyses assessing (1) the effects of physician profile characteristics (i.e., race, age, and gender) on African American women's perception of physician trustworthiness and (2) severity of visit (routine versus serious health concern visit) on women's perceptions of physician trustworthiness. After adjusting for all study covariates, there were no significant differences in overall (or index of) trust by physician profile characteristics. However, there were significant differences by physician profile characteristics on the individual measures of fidelity, competence and honesty. White-older-male when compared to AA-older-female were rated more favorably on fidelity (4.23, 4.02; ES = 0.215, 95% CI: 0.001-0.431), competence (4.23, 3.95; ES = 0.278, 95% CI: 0.062-0.494) and honesty (4.39, 4.19, ES = 0.215, 95% CI: 0.001-0.431). AA-older-male were rated more favorably than AA-older-female on competence (4.20, 3.95; ES = 0.243, 95% CI: 0.022-0.464) and honesty (4.44, 4.19; ES = 0.243, 95% CI: 0.022-0.464). AA-young-male was rated more favorably than AA-older-female on competence (4.16, 3.95; ES = 0.205, 95% CI: 0.013-0.423). There were no significant differences in perceptions of trust by type of medical visit (results not shown). Higher education was associated with less favorable scores on fidelity, honesty, global trust, and overall trust index (results not shown).
% (N) or mean (SD) | |
Age | |
45-64 | 73.2 (229) |
65+ | 26.8 (84) |
Education | |
< High school | 18.2 (57) |
High school | 23.0 (72) |
Some college | 31.0 (97) |
College + | 27.8 (87) |
Income | |
< $25,000 | 45.7 (143) |
$25,000-$49, 999 | 30.7 (96) |
$50,000-$74, 999 | 13.4 (42) |
$75,000 or more | 10.2 (32) |
No health insurance | 16.0 (50) |
No Usual provider | 6.4 (20) |
Self-reported health | |
Poor | 7.4 (23) |
Fair | 24.3 (76) |
Good | 43.7 (137) |
Very good | 20.4 (64) |
Excellent | 4.2 (13) |
Negative experience with health provider | 43.8 (176) |
Trust measures |
|
Fidelity | 3.82 (1.004) |
Competence | 3.86 (0.986) |
Honesty | 3.96 (0.993) |
Confidentiality | 4.07 (1.012) |
Global trust | 3.85 (1.041) |
Trust index score |
3.91 (1.007) |
*Scores are based on a scale of 1 to 5, with higher scores indicating higher trust. †Five trust measures were summed and averaged. |
*Doctor Profile Abbreviations: AAYM: African American Young Male; AAOM: African American Older Male; AAYF: African American Young Female; AAOF: African American Older Female; WYM: White Young Male; WOM: White Older Male; WYF: White Young Female; WOF: White Older Female.†All profiles were compared to AAOF. Effect size: d = 0.1 (small effect), d = 0.3 (medium effect), d = 0.5 (large effect).
In this study, African American women were given medical visit vignettes and pictures of imaginary pictures physicians (in which race/ethnicity, age, and gender were randomly manipulated) and asked to make judgments about perceived fidelity, competence, honesty, confidentiality, and global trust of the physician. The index of trust (five measures were summed and averaged) did not differ significantly by physician characteristics. However, there were small statistically significant differences by physician characteristics on the measures of fidelity, honesty, and competence. The White-older-male was rated more favorably than the AA-older-female on fidelity. The White-older-male and the AA-older-male were rated more favorably than the AA-older-female on honesty. The White-older-male, AA-older-male, and AA-young-male were rated more favorably than AA-older-female on competence. Type of medical visit did not influence perceptions of physician trustworthiness.
Our finding of no difference in overall trust by physician characteristics suggest that our sample of African Americans may not have physician preferences or it may be that they had professed overall trust for physicians irrespective of race/ethnicity, gender and age. This is largely consistent with the reviews on patient-physician race concordance, which concluded that the majority of patients have no preference regarding providers' race/ethnicity and are very satisfied with the care they receive from physicians of dissimilar race/ethnicity [14,15]. However, this finding is more in line with Street and colleagues (2008) study which showed that patients perceptions of shared identity of race, ethnicity, and community with physicians was not associated with trust; whereas perceptions of shared personal believe, values and ways of communicating were associated with higher ratings of trust in physicians [6]. Street and colleagues (2008) also found that some patients in race and gender concordant dyads perceive themselves dissimilar from their physicians, while others in discordant dyads perceive themselves as similar to their physicians [6]. On balance, these data represent aggregates; individual differences are bound to exist.
On the other hand, our slightly higher ratings for older male physicians compared to older female African American physicians on selected measures of trust may refer to medical setting, societal beliefs, patient values and expectations [1,40]. Kumar and colleagues (2009) found that race concordance or discordance with a physician was largely a function of the medical setting [41]. In settings that employ a large number of African American physicians, Whites with health insurance are more likely to be in concordant dyads than Whites without insurance and African Americans without insurance were more likely to be in a concordant dyad than African Americans with insurance [41]. Race concordant or discordant dyads did not influence patients' perceived quality of care [41]. There are situations, context, and settings that shape patients' perceptions of providers and healthcare; thus, it is critical that we do not over generalize the importance of concordance to patients.
Gender roles and beliefs are pervasive in society, in many professional contexts women's work roles and competences are more likely to be devalued [42]. Gender differences favoring men also exist in patients' evaluation of clinical performance: many patients do not give female physicians the credit they deserve [40]. Research shows that high verbal patient-centered behavior is regarded as an indicator of clinical competence for male physicians but not female physicians [21]. Female physicians have a more patient-centered practice style which is preferred by patients and is associated with higher patient satisfaction [40,43]. However, the meta-analysis of patient satisfaction and physician gender conducted by Hall and colleagues found an almost nonexistent difference favoring female physicians on care satisfaction [43]. Female physicians were slightly favored when physicians were less experienced when satisfaction pertained to a specific visit, and when patients were young [43]. Relatedly, the slight preference for older male physicians (on fidelity, honesty, and competence) by the older African American women in our study may be due to implicit or unconscious bias [21].
We did not demonstrate a relationship between physician characteristics and perceived trustworthiness of physicians, which may speak to the complexity of concordance, trust and the patient-provider relationships that have been documented [1,6,14]. A gestalt of patient, provider and system factors may determine patient trust and preference for concordance [1,14,15]. While concordance may orient patients and physicians towards some common ground [6], a physician's character and personality in an ongoing patient-physician relationship impacts a patient's trust in that physician [1].
Our results should be considered in the context of several limitations. First, we used a convenience sample of women which precludes generalizability to other African American populations. Second, there is evidence of discrepancies between social judgment in vignette surveys and actual behavior [27]. Third, every aspect of a social phenomenon cannot be simulated and it is also difficult to assess the extent to which social desirability influenced the subjects' responses [24]. Finally, this study used observational methods and therefore cannot define cause-and-effect associations. However, with the paucity of research on trust among older African Americans [26] and the ambiguity regarding patient preferences for concordance with their physicians [14,15], this research fills a gap in the literature and takes a novel approach to understanding how older African American women may make choices and judgments about trusting their physicians.
Our study has implications for programs and policies focused on reducing racial/ethnic disparities by promoting diversity in the healthcare workforce. Our findings suggest that concordance may hold no salience for some groups of older African women. The relevance of race/ethnic concordance has been questioned because it does not account for multiracial identity and the variability in values, beliefs, and culture within racial/ethnic groups [14]. Moreover, it might create the perception that health equity can only be achieved in race-concordant patient-physician, which is unrealistic given our multiracial identity and cultures [14]. Promoting diversity in the medical profession is a laudable goal that will address an equity issue [44]; however, we should also strive to create a culturally competency diverse healthcare workforce. While diversity may reduce disparities in access because ethnic minority physicians are more likely to serve disadvantaged populations [15], training culturally competent physicians is paramount in reducing disparities in health outcomes [6,15,44]. Cultural competency can foster trust and communication, which are critical to the patient-provider relationship and to improving disparities in health outcomes [1,6,15]. Cultural competency training can transcend concordance issues to establish a positive relationship with a patient, which contributes to positive health outcomes [6].
This project was supported by the NIH Loan Repayment Program and the Clinical and Translational Science Award (CTSA) program, through the NIH National Center for Advancing Translational Sciences (NCATS), grant UL1TR000427. Preliminary findings of this project were present at the 2010 Annual Gerontological Society of America meeting.
The authors have no potential conflicts of interests, financial or otherwise.
[1] | Coley WB (1893) The Treatment of Malignant Tumors by Repeated Innoculations of Erysipelas: With a Report of Ten Original Cases. Am J Med Sci 10: 487-511. |
[2] | Pick TP (1899) Surgery: Green. 1208 p. |
[3] |
Menzies SW, McCarthy WH (1997) Complete regression of primary cutaneous malignant melanoma. Arch Surg 132: 553-556. doi: 10.1001/archsurg.1997.01430290099020
![]() |
[4] |
Quaglino P, Marenco F, Osella-Abate S, et al. (2010) Vitiligo is an independent favourable prognostic factor in stage III and IV metastatic melanoma patients: results from a single-institution hospital-based observational cohort study. Ann Onc 21: 409-414. doi: 10.1093/annonc/mdp325
![]() |
[5] |
Topalian SL, Sznol M, McDermott DF, et al. (2014) Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Onc 32:1020-1030. doi: 10.1200/JCO.2013.53.0105
![]() |
[6] |
Rosenberg SA, Yang JC, Sherry RM, et al. (2011) Durable Complete Responses in Heavily Pretreated Patients with Metastatic Melanoma Using T-Cell Transfer Immunotherapy. Clin Cancer Res 17: 4550-4557. doi: 10.1158/1078-0432.CCR-11-0116
![]() |
[7] |
McDermott D, Lebbé|C, Hodi FS, et al. (2014) Durable benefit and the potential for long-term survival with immunotherapy in advanced melanoma. Cancer Treat Rev 40: 1056-1064. doi: 10.1016/j.ctrv.2014.06.012
![]() |
[8] |
Sanlorenzo M, Vujic I, Posch C, et al. (2014) Melanoma immunotherapy. Cancer Biol Ther 15:665-674. doi: 10.4161/cbt.28555
![]() |
[9] | Avril MF, Aamdal S, Grob JJ, et al. (2004) Fotemustine compared with dacarbazine in patients with disseminated malignant melanoma: a phase III study. J Clin Onc.22: 1118-1125. |
[10] |
Garbe C, Eigentler TK, Keilholz U, et al. (2011) Systematic Review of Medical Treatment in Melanoma: Current Status and Future Prospects. Oncologist 16: 5-24. doi: 10.1634/theoncologist.2010-0190
![]() |
[11] |
Hervas-Stubbs S, Perez-Gracia JL, Rouzaut A, et al. (2011) Direct effects of type I interferons on cells of the immune system. Clin Cancer Res 17: 2619-2627. doi: 10.1158/1078-0432.CCR-10-1114
![]() |
[12] | Kirkwood JM, Strawderman MH, Ernstoff MS, et al. (1996) Interferon alfa-2b adjuvant therapy of high-risk resected cutaneous melanoma: the Eastern Cooperative Oncology Group Trial EST1684. J Clin Onc 14: 7-17. |
[13] |
Payne MJ, Argyropoulou K, Lorigan P, et al. (2014) Phase II Pilot Study of Intravenous High-Dose Interferon With or Without Maintenance Treatment in Melanoma at High Risk of Recurrence. J Clin Onc 32: 185-190. doi: 10.1200/JCO.2013.49.8717
![]() |
[14] | Di Trolio R, Simeone E, Di Lorenzo G, et al. (2014) The use of interferon in melanoma patients: A systematic review. Cytokine Growth Factor Rev Epub |
[15] |
Kaufman HL, Kirkwood JM, Hodi FS, et al. (2013) The Society for Immunotherapy of Cancer consensus statement on tumour immunotherapy for the treatment of cutaneous melanoma. Nat Rev Clin Onc 10: 588-598. doi: 10.1038/nrclinonc.2013.153
![]() |
[16] | Baker DE (2001) Pegylated interferons. Rev Gastroenterol Disord 1: 87-99. |
[17] |
Eggermont AMM, Suciu S, Testori A, et al. (2012) Ulceration and stage are predictive of interferon efficacy in melanoma: results of the phase III adjuvant trials EORTC 18952 and EORTC 18991. Eur J Cancer 48: 218-225. doi: 10.1016/j.ejca.2011.09.028
![]() |
[18] |
Hofmann MA, Kiecker F, Küchler I, et al. (2011) Serum TNF-α, B2M and sIL-2R levels are biological correlates of outcome in adjuvant IFN-α2b treatment of patients with melanoma. J Cancer Res Clin Onc 137: 455-462. doi: 10.1007/s00432-010-0900-1
![]() |
[19] |
Flaherty LE, Othus M, Atkins MB, et al. (2014) Southwest Oncology Group S0008: A Phase III Trial of High-Dose Interferon Alfa-2b Versus Cisplatin, Vinblastine, and Dacarbazine, Plus Interleukin-2 and Interferon in Patients With High-Risk Melanoma-An Intergroup Study of Cancer and Leukemia Group B, Children's Oncology Group, Eastern Cooperative Oncology Group, and Southwest Oncology Group. J Clin Onc 32: 3771-3778. doi: 10.1200/JCO.2013.53.1590
![]() |
[20] |
Kim KB, Legha SS, Gonzalez R, et al. (2009) A randomized phase III trial of biochemotherapy versus interferon-alpha-2b for adjuvant therapy in patients at high risk for melanoma recurrence. Melanoma Res 19: 42-49. doi: 10.1097/CMR.0b013e328314b84a
![]() |
[21] | Atkins MB, Kunkel L, Sznol M, et al. (2000) High-dose recombinant interleukin-2 therapy in patients with metastatic melanoma: long-term survival update. Cancer J Sc Am 6: S11-14. |
[22] |
Schwartzentruber DJ (2001) Guidelines for the safe administration of high-dose interleukin-2. J Immunotherapy (Hagerstown, Md: 1997) 24: 287-293. doi: 10.1097/00002371-200107000-00004
![]() |
[23] |
Petrella T, Quirt I, Verma S, et al. (2007) Single-agent interleukin-2 in the treatment of metastatic melanoma: A systematic review. Cancer Treat Rev 33: 484-496. doi: 10.1016/j.ctrv.2007.04.003
![]() |
[24] |
Joseph RW, Sullivan RJ, Harrell R, et al. (2012) Correlation of NRAS Mutations With Clinical Response to High-dose IL-2 in Patients With Advanced Melanoma. J Immunotherapy 35: 66-72. doi: 10.1097/CJI.0b013e3182372636
![]() |
[25] | Keilholz U, Conradt C, Legha SS, et al. (1998) Results of interleukin-2-based treatment in advanced melanoma: a case record-based analysis of 631 patients. J Clin Onc 16: 2921-2929. |
[26] |
Byers BA, Temple-Oberle CF, Hurdle V, et al. (2014) Treatment of in-transit melanoma with intra-lesional interleukin-2: A systematic review. J Surg Onc 110: 770-775. doi: 10.1002/jso.23702
![]() |
[27] |
Boyd KU, Wehrli BM, Temple CLF (2011) Intra-lesional interleukin-2 for the treatment of in-transit melanoma. J Surg Onc 104: 711-717. doi: 10.1002/jso.21968
![]() |
[28] | Daud AI, DeConti RC, Andrews S, et al. (2008) Phase I trial of interleukin-12 plasmid electroporation in patients with metastatic melanoma. J Clin Onc 26: 5896-5903. |
[29] |
Dillman RO, Cornforth AN, Depriest C, et al. (2012) Tumor stem cell antigens as consolidative active specific immunotherapy: a randomized phase II trial of dendritic cells versus tumor cells in patients with metastatic melanoma. J Immunotherapy 35: 641-649. doi: 10.1097/CJI.0b013e31826f79c8
![]() |
[30] |
Dillman RO, DePriest C, Ellis RA (2014) Long-term survival for patients with detectable metastatic melanoma at time of treatment with patient-specific tumor stem cell vaccines. J Clin Onc 32: 5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[31] |
Dudley ME, Yang JC, Sherry R, et al. (2008) Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Onc 26: 5233-5239. doi: 10.1200/JCO.2008.16.5449
![]() |
[32] |
Radvanyi LG, Bernatchez C, Zhang M, et al. (2012) Specific lymphocyte subsets predict response to adoptive cell therapy using expanded autologous tumor-infiltrating lymphocytes in metastatic melanoma patients. Clin Cancer Res 18: 6758-6770. doi: 10.1158/1078-0432.CCR-12-1177
![]() |
[33] |
Besser MJ, Shapira-Frommer R, Treves AJ, et al. (2010) Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin Cancer Res 16: 2646-2655. doi: 10.1158/1078-0432.CCR-10-0041
![]() |
[34] |
Collins AV, Brodie DW, Gilbert RJC, et al. (2002) The interaction properties of costimulatory molecules revisited. Immunity 17: 201-210. doi: 10.1016/S1074-7613(02)00362-X
![]() |
[35] |
Hodi FS, O'Day SJ, McDermott DF, et al. (2010) Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363: 711-723. doi: 10.1056/NEJMoa1003466
![]() |
[36] |
Wolchok JD, Neyns B, Linette G, et al. (2010) Ipilimumab monotherapy in patients with pretreated advanced melanoma: a randomised, double-blind, multicentre, phase 2, dose-ranging study. Lancet Onc 11: 155-164. doi: 10.1016/S1470-2045(09)70334-1
![]() |
[37] |
Robert C, Thomas L, Bondarenko I, et al. (2011) Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med 364: 2517-2526. doi: 10.1056/NEJMoa1104621
![]() |
[38] |
Lebbé|C, Weber JS, Maio M, et al. (2014) Survival follow-up and ipilimumab retreatment of patients with advanced melanoma who received ipilimumab in prior phase II studies. Ann Onc/ESMO 25: 2277-2284. doi: 10.1093/annonc/mdu441
![]() |
[39] | Schadendorf D, FS Hodi, C Robert, et al. (2013) Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in metastatic or locally advanced, unresectable melanoma. ESMO 2013 Congress. |
[40] |
Chiarion-Sileni V, Pigozzo J, Ascierto PA, et al. (2014) Ipilimumab retreatment in patients with pretreated advanced melanoma: the expanded access programme in Italy. Br J Cancer 110:1721-1726. doi: 10.1038/bjc.2014.126
![]() |
[41] |
Dequen P, Lorigan P, Jansen JP, et al. (2012) Systematic review and network meta-analysis of overall survival comparing 3 mg/kg ipilimumab with alternative therapies in the management of pretreated patients with unresectable stage III or IV melanoma. Oncologist 17: 1376-1385. doi: 10.1634/theoncologist.2011-0427
![]() |
[42] |
Ascierto PA, Simeone E, Sileni VC, et al. (2014) Sequential treatment with ipilimumab and BRAF inhibitors in patients with metastatic melanoma: data from the Italian cohort of the ipilimumab expanded access program. Cancer Inves. 32: 144-149. doi: 10.3109/07357907.2014.885984
![]() |
[43] |
Hodi FS, Lee S, McDermott DF, et al. (2014) Ipilimumab plus sargramostim vs ipilimumab alone for treatment of metastatic melanoma: A randomized clinical trial. JAMA 312: 1744-1753. doi: 10.1001/jama.2014.13943
![]() |
[44] |
Bapodra A, Silva IEDPD, Lui KP, et al. (2014) Clinical outcome and CD4+ differentiation in anti-CTLA-4/radiation and anti-CTLA-4/steroid therapy. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[45] |
Downey SG, Klapper JA, Smith FO, et al. (2007) Prognostic factors related to clinical response in patients with metastatic melanoma treated by CTL-associated antigen-4 blockade. Clin Cancer Res 13: 6681-6688. doi: 10.1158/1078-0432.CCR-07-0187
![]() |
[46] |
Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. (2014) Ipilimumab versus placebo after complete resection of stage III melanoma: Initial efficacy and safety results from the EORTC 18071 phase III trial. J Clin Onc 32: 5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[47] |
Hamid O, Schmidt H, Nissan A, et al. (2011) A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J Transl Med 9: 204. doi: 10.1186/1479-5876-9-204
![]() |
[48] |
Adaniel C, Rendleman J, Polsky D, et al. (2014) Germline genetic determinants of immunotherapy response in metastatic melanoma. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[49] |
Freeman-Keller M, Weber JS (2015) Anti-programmed death receptor 1 immunotherapy in melanoma: rationale, evidence and clinical potential. Ther Adv Med Onc 7: 12-21. doi: 10.1177/1758834014551747
![]() |
[50] | Weber JS, Minor DR, D'Angelo S, et al. (2014) A Phase 3 randomized, open-label study of nivolumab (anti-PD-1|BMS-936558|ONO-4538) versus investigator's choice chemotherapy (ICC) in patients with advanced melanoma after prior anti-CTLA-4 therapy. ESMO 2014 Congress. |
[51] |
Hamid O, Robert C, Daud A, et al. (2013) Safety and tumor responses with lambrolizumab (anti-PD-1) in melanoma. N Engl J Med 369: 134-144. doi: 10.1056/NEJMoa1305133
![]() |
[52] |
Robert C, Ribas A, Wolchok JD, et al. (2014) Anti-programmed-death-receptor-1 treatment with pembrolizumab in ipilimumab-refractory advanced melanoma: a randomised dose-comparison cohort of a phase 1 trial. The Lancet 384: 1109-1117. doi: 10.1016/S0140-6736(14)60958-2
![]() |
[53] | Robert C, Joshua AM, Weber AS (2014) Pembrolizumab (pembro|MK-3475) for advanced melanoma (MEL): Randomized comparison of two dosing schedules. ESMO 2014 Congress. |
[54] |
Ribas A, Hodi FS, Kefford R, et al. (2014) Efficacy and safety of the anti-PD-1 monoclonal antibody MK-3475 in 411 patients (pts) with melanoma (MEL). J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[55] |
Kefford R, Ribas A, Hamid O, et al. (2014) Clinical efficacy and correlation with tumor PD-L1 expression in patients (pts) with melanoma (MEL) treated with the anti-PD-1 monoclonal antibody MK-3475. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[56] |
Wolchok JD, Kluger H, Callahan MK, et al. (2013) Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med 369: 122-133. doi: 10.1056/NEJMoa1302369
![]() |
[57] | Sharabi AB, Nirschl CJ, Kochel CM, et al. (2014) Stereotactic Radiation Therapy Augments Antigen-Specific PD-1 Mediated Anti-Tumor Immune Responses via Cross-Presentation of Tumor Antigen. Cancer Immunol Res.: canimm.0196.2014. |
[58] |
Brahmer JR, Tykodi SS, Chow LQM, et al. (2012) Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med 366: 2455-2465. doi: 10.1056/NEJMoa1200694
![]() |
[59] |
Fonsatti E, Maio M, Altomonte M, et al. (2010) Biology and Clinical Applications of CD40 in Cancer Treatment. Seminars Onc 37: 517-523. doi: 10.1053/j.seminoncol.2010.09.002
![]() |
[60] |
Vonderheide RH, Flaherty KT, Khalil M, et al. (2007) Clinical activity and immune modulation in cancer patients treated with CP-870,893, a novel CD40 agonist monoclonal antibody. J Clin Onc 25: 876-883. doi: 10.1200/JCO.2006.08.3311
![]() |
[61] |
Hemon P, Jean-Louis F, Ramgolam K, et al. (2011) MHC class II engagement by its ligand LAG-3 (CD223) contributes to melanoma resistance to apoptosis. J Immunol 186: 5173-5183. doi: 10.4049/jimmunol.1002050
![]() |
[62] |
Baitsch L, Baumgaertner P, Devêvre E, et al. (2011) Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J Clin Invest 121: 2350-2360. doi: 10.1172/JCI46102
![]() |
[63] |
da Silva IP, Gallois A, Jimenez-Baranda S, et al. (2014) Reversal of NK-cell exhaustion in advanced melanoma by Tim-3 blockade. Cancer Immunol Res 2: 410-422. doi: 10.1158/2326-6066.CIR-13-0171
![]() |
[64] | Lombardi VC, Khaiboullina SF, Rizvanov AA (2015) Plasmacytoid dendritic cells, a role in neoplastic prevention and progression. Eur J Clin Invest 45: 1-8. |
[65] |
Aspord C, Tramcourt L, Leloup C, et al. (2014) Imiquimod inhibits melanoma development by promoting pDC cytotoxic functions and impeding tumor vascularization. J Invest Derm 134:2551-2561. doi: 10.1038/jid.2014.194
![]() |
[66] | Hyde MA, Hadley ML, Tristani-Firouzi P, et al. (2012) A randomized trial of the off-label use of imiquimod, 5%, cream with vs without tazarotene, 0.1%, gel for the treatment of lentigo maligna, followed by conservative staged excisions. Arch Derm 148: 592-596. |
[67] |
Singh M, Khong H, Dai Z, et al. (2014) Effective innate and adaptive antimelanoma immunity through localized TLR7/8 activation. J Immunol 193: 4722-4731. doi: 10.4049/jimmunol.1401160
![]() |
[68] | Andtbacka RHI, Collichio FA, Amatruda T, et al. (2013) OPTiM: A randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. J Clin Onc 31. |
[69] |
Andtbacka RHI, Curti BD, Kaufman H, et al. (2014) CALM study: A phase II study of an intratumorally delivered oncolytic immunotherapeutic agent, coxsackievirus A21, in patients with stage IIIc and stage IV malignant melanoma. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[70] |
Zamarin D, Holmgaard RB, Subudhi SK, et al. (2014) Potentiation of immune checkpoint blockade cancer immunotherapy with oncolytic virus. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[71] | Schipper H, Alla V, Meier C, et al. (2014) Eradication of metastatic melanoma through cooperative expression of RNA-based HDAC1 inhibitor and p73 by oncolytic adenovirus. Oncotarget 5: 5893-5907. |
[72] |
Jemal A, Bray F, Center MM, et al. (2011) Global cancer statistics. CA Cancer J Clin 61: 69-90. doi: 10.3322/caac.20107
![]() |
[73] |
74. Kono K, Mizukami Y, Daigo Y, et al. (2009) Vaccination with multiple peptides derived from novel cancer-testis antigens can induce specific T-cell responses and clinical responses in advanced esophageal cancer. Cancer Sci 100: 1502-1509. doi: 10.1111/j.1349-7006.2009.01200.x
![]() |
[74] |
75. Iinuma H, Fukushima R, Inaba T, et al. (2014) Phase I clinical study of multiple epitope peptide vaccine combined with chemoradiation therapy in esophageal cancer patients. J Transl Med 12:84. doi: 10.1186/1479-5876-12-84
![]() |
[75] | 76. Toh U, Yamana H, Sueyoshi S, et al. (2000) Locoregional cellular immunotherapy for patients with advanced esophageal cancer. Clin Cancer Res 6: 4663-4673. |
[76] | 77. Akutsu Y, Qin W, Murakami K, et al. (2014) The effect of stress on efficacy of dendritic cell therapy for esophageal squamous cell carcinoma. J Clin Onc32. |
[77] | 79. Crew KD, Neugut AI (2006) Epidemiology of gastric cancer. World J Gastroenterol 12:354-362. |
[78] |
80. Ohtsu A, Shah MA, Van Cutsem E, et al. (2011) Bevacizumab in combination with chemotherapy as first-line therapy in advanced gastric cancer: a randomized, double-blind, placebo-controlled phase III study. J Clin Onc 29: 3968-3976. doi: 10.1200/JCO.2011.36.2236
![]() |
[79] |
81. Bang Y-J, Van Cutsem E, Feyereislova A, et al. (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376: 687-697. doi: 10.1016/S0140-6736(10)61121-X
![]() |
[80] |
82. Ajani JA, Hecht JR, Ho L, et al. (2006) An open-label, multinational, multicenter study of G17DT vaccination combined with cisplatin and 5-fluorouracil in patients with untreated,advanced gastric or gastroesophageal cancer: the GC4 study. Cancer 106: 1908-1916. doi: 10.1002/cncr.21814
![]() |
[81] | 83. Masuzawa T, Fujiwara Y, Okada K, et al. (2012) Phase I/II study of S-1 plus cisplatin combined with peptide vaccines for human vascular endothelial growth factor receptor 1 and 2 in patients with advanced gastric cancer. Int J Onc 41: 1297-1304. |
[82] |
84. Popiela T, Kulig J, Czupryna A, et al. (2004) Efficiency of adjuvant immunochemotherapy following curative resection in patients with locally advanced gastric cancer. Gastric Cancer 7:240-245. doi: 10.1007/s10120-004-0299-y
![]() |
[83] | 85. Kono K, Takahashi A, Sugai H, et al. (2002) Dendritic cells pulsed with HER-2/neu-derived peptides can induce specific T-cell responses in patients with gastric cancer. Clin Cancer Res 8:3394-3400. |
[84] | 86. Kono K, Takahashi A, Ichihara F, et al. (2002) Prognostic significance of adoptive immunotherapy with tumor-associated lymphocytes in patients with advanced gastric cancer: a randomized trial. Clin Cancer Res 8: 1767-1771. |
[85] |
87. Jiang J-T, Shen Y-P, Wu C-P, et al. (2010) Increasing the frequency of CIK cells adoptive immunotherapy may decrease risk of death in gastric cancer patients. World J Gastroenterol 16:6155-6162. doi: 10.3748/wjg.v16.i48.6155
![]() |
[86] |
88. Shi L, Zhou Q, Wu J, et al. (2012) Efficacy of adjuvant immunotherapy with cytokine-induced killer cells in patients with locally advanced gastric cancer. Cancer Immunol. Immunotherapy 61:2251-2259. doi: 10.1007/s00262-012-1289-2
![]() |
[87] | 89. Jeung H-C, Moon YW, Rha SY, et al. (2008) Phase III trial of adjuvant 5-fluorouracil and adriamycin versus 5-fluorouracil, adriamycin, and polyadenylic-polyuridylic acid (poly A:U) for locally advanced gastric cancer after curative surgery: final results of 15-year follow-up. Ann Onc/ESMO 19: 520-526. |
[88] | 90. Muro K, Bang Y, Shankaran V, et al. (2014) A phase 1b study of pembrolizumab (Pembro|MK-3475) in patients (Pts) with advanced gastric cancer. ESMO 2014 Congress. |
[89] |
91. Diermeier-Daucher S, Ortmann O, Buchholz S, et al. (2012) Trifunctional antibody ertumaxomab. mAbs 4: 614-622. doi: 10.4161/mabs.21003
![]() |
[90] |
92. Haense N, Pauligk C, Marme F, et al. (2014) Interim analysis of a phase I/II open label, dose-escalating study to investigate safety, tolerability, and preliminary efficacy of the trifunctional anti-HER2/neu x anti-CD3 antibody ertumaxomab in patients with HER2/neu expressing solid tumors progressing after standard therapy. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[91] |
93. Atanackovic D, Reinhard H, Meyer S, et al. (2013) The trifunctional antibody catumaxomab amplifies and shapes tumor-specific immunity when applied to gastric cancer patients in the adjuvant setting. Hum Vaccines Immunotherapeutics 9: 2533-2542. doi: 10.4161/hv.26065
![]() |
[92] |
94. Heiss MM, Murawa P, Koralewski P, et al. (2010) The trifunctional antibody catumaxomab for the treatment of malignant ascites due to epithelial cancer: Results of a prospective randomized phase II/III trial. Int J Cancer 127: 2209-2221. doi: 10.1002/ijc.25423
![]() |
[93] |
95. Matsueda S (2014) Immunotherapy in gastric cancer. World J Gastroenterol 20: 1657. doi: 10.3748/wjg.v20.i7.1657
![]() |
[94] |
96. Gabitass RF, Annels NE, Stocken DD, et al. (2011) Elevated myeloid-derived suppressor cells in pancreatic, esophageal and gastric cancer are an independent prognostic factor and are associated with significant elevation of the Th2 cytokine interleukin-13. Cancer Immunol Immunother 60: 1419-1430. doi: 10.1007/s00262-011-1028-0
![]() |
[95] |
97. Mundy-Bosse BL, Young GS, Bauer T, et al. (2011) Distinct myeloid suppressor cell subsets correlate with plasma IL-6 and IL-10 and reduced interferon-alpha signaling in CD4+ T cells from patients with GI malignancy. Cancer Immunol Immunother: CII 60: 1269-1279. doi: 10.1007/s00262-011-1029-z
![]() |
[96] | 98. Jaffee E (2014) Cancer Research Institute: Pancreatic Cancer. Available from: http://www.cancerresearch.org/cancer-immunotherapy/impacting-all-cancers/pancreatic-cancer |
[97] | 99. Toomey PG, Vohra NA, Ghansah T, et al. (2013) Immunotherapy for gastrointestinal malignancies. Cancer Control 20: 32-42. |
[98] |
100. Uram JN, Le DT (2013) Current advances in immunotherapy for pancreatic cancer. Curr. Problems Cancer 37: 273-279. doi: 10.1016/j.currproblcancer.2013.10.004
![]() |
[99] | 101. Lepisto AJ, Moser AJ, Zeh H, et al. (2008) A phase I/II study of a MUC1 peptide pulsed autologous dendritic cell vaccine as adjuvant therapy in patients with resected pancreatic and biliary tumors. Cancer Ther. 6: 955-964. |
[100] |
102. Bernhardt SL, Gjertsen MK, Trachsel S, et al. (2006) Telomerase peptide vaccination of patients with non-resectable pancreatic cancer: a dose escalating phase I/II study. Br J Cancer 95:1474-1482. doi: 10.1038/sj.bjc.6603437
![]() |
[101] |
103. Salman B, Zhou D, Jaffee EM, et al. (2013) Vaccine therapy for pancreatic cancer. Oncoimmunology 2: e26662. doi: 10.4161/onci.26662
![]() |
[102] | 104. Middleton GW, Valle JW, Wadsley J, et al. (2013) A phase III randomized trial of chemoimmunotherapy comprising gemcitabine and capecitabine with or without telomerase vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer. J Clin Onc 31. |
[103] |
105. Asahara S, Takeda K, Yamao K, et al. (2013) Phase I/II clinical trial using HLA-A24-restricted peptide vaccine derived from KIF20A for patients with advanced pancreatic cancer. J Transl Med 11: 291. doi: 10.1186/1479-5876-11-291
![]() |
[104] |
106. Gjertsen MK, Buanes T, Rosseland AR, et al. (2001) Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: Clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer 92: 441-450. doi: 10.1002/ijc.1205
![]() |
[105] |
107. Wedén S, Klemp M, Gladhaug IP, et al. (2011) Long-term follow-up of patients with resected pancreatic cancer following vaccination against mutant K-ras. Int J Cancer 128: 1120-1128. doi: 10.1002/ijc.25449
![]() |
[106] | 108. Jaffee EM, Hruban RH, Biedrzycki B, et al. (2001) Novel allogeneic granulocyte-macrophage colony-stimulating factor-secreting tumor vaccine for pancreatic cancer: a phase I trial of safety and immune activation. J Clin Onc 19: 145-156. |
[107] |
109. Lutz E, Yeo CJ, Lillemoe KD, et al. (2011) A Lethally Irradiated Allogeneic Granulocyte-Macrophage Colony Stimulating Factor-Secreting Tumor Vaccine for Pancreatic Adenocarcinoma: A Phase II Trial of Safety, Efficacy, and Immune Activation. Ann Surg 253:328-335. doi: 10.1097/SLA.0b013e3181fd271c
![]() |
[108] | 110. Le DT, Wang-Gillam A, Picozzi V, et al. (2014) A phase 2, randomized trial of GVAX pancreas and CRS-207 immunotherapy versus GVAX alone in patients with metastatic pancreatic adenocarcinoma: Updated results. J Clin Onc 32. |
[109] |
111. Rossi GR, Lima CMSR, Hardacre JM, et al. (2014) Correlation of anti-calreticulin antibody titers with improved overall survival in a phase 2 clinical trial of algenpantucel-L immunotherapy for patients with resected pancreatic cancer. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[110] | 112. Starodub A, Ocean AJ, Messersmith WA, et al. (2015) Phase I/II trial of IMMU-132 (isactuzumab govitecan), an anti-Trop-2-SN-38 antibody drug conjugate (ADC): Results in patients with metastatic gastrointestinal (GI) cancers. J Clin Onc 33. |
[111] | 113. Kondo H, Hazama S, Kawaoka T, et al. (2008) Adoptive immunotherapy for pancreatic cancer using MUC1 peptide-pulsed dendritic cells and activated T lymphocytes. Anticancer Res 28:379-387. |
[112] |
114. Chung MJ, Park JY, Bang S, et al. (2014) Phase II clinical trial of ex vivo-expanded cytokine-induced killer cells therapy in advanced pancreatic cancer. Cancer Immunol Immunother 63: 939-946. doi: 10.1007/s00262-014-1566-3
![]() |
[113] |
115. Royal RE, Levy C, Turner K, et al. (2010) Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunotherapy (Hagerstown, Md: 1997) 33: 828-833. doi: 10.1097/CJI.0b013e3181eec14c
![]() |
[114] | 116. Le DT, Lutz E, Uram JN, et al. (2013) Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J Immunotherapy (Hagerstown, Md: 1997) 36: 382-389. |
[115] |
117. Beatty GL, Chiorean EG, Fishman MP, et al. (2011) CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 331: 1612-1616. doi: 10.1126/science.1198443
![]() |
[116] |
118. Pernot S, Terme M, Voron T, et al. (2014) Colorectal cancer and immunity: what we know and perspectives. World J Gastroenterol 20: 3738-3750. doi: 10.3748/wjg.v20.i14.3738
![]() |
[117] |
120. Staff C, Mozaffari F, Haller BK, et al. (2011) A Phase I safety study of plasmid DNA immunization targeting carcinoembryonic antigen in colorectal cancer patients. Vaccine 29:6817-6822. doi: 10.1016/j.vaccine.2010.12.063
![]() |
[118] |
121. Albanopoulos K, Armakolas A, Konstadoulakis MM, et al. (2000) Prognostic significance of circulating antibodies against carcinoembryonic antigen (anti-CEA) in patients with colon cancer. Am J Gastroenterol 95: 1056-1061. doi: 10.1111/j.1572-0241.2000.01982.x
![]() |
[119] | 122. Conry RM, Allen KO, Lee S, et al. (2000) Human autoantibodies to carcinoembryonic antigen (CEA) induced by a vaccinia-CEA vaccine. Clin Cancer Res 6: 34-41. |
[120] | 123. Loibner H, Eckert H, Eller N (2004) A randomized placebo-controlled phase II study with the cancer vaccine IGN101 in patients with epithelial solid organ tumors (IGN101/2-01). J Clin Oncol 22: 2619. |
[121] |
124. Karanikas V, Thynne G, Mitchell P, et al. (2001) Mannan Mucin-1 Peptide Immunization: Influence of Cyclophosphamide and the Route of Injection. J Immunotherapy 24: 172-183. doi: 10.1097/00002371-200103000-00012
![]() |
[122] | 125. Hazama S, Nakamura Y, Takenouchi H, et al. (2014) A phase I study of combination vaccine treatment of five therapeutic epitope-peptides for metastatic colorectal cancer|safety, immunological response, and clinical outcome. J Transl Med 12: 63. |
[123] | 126. Ibrahim R, Achtar M, Herrin V (2004) Mutant P53 vaccination of patients with advanced cancers generates specific immunological responses. J Clin Oncol 22: 2521. |
[124] |
127. Toubaji A, Achtar M, Provenzano M, et al. (2008) Pilot study of mutant ras peptide-based vaccine as an adjuvant treatment in pancreatic and colorectal cancers. Cancer Immunol Immunother 57: 1413-1420. doi: 10.1007/s00262-008-0477-6
![]() |
[125] | 128. Harris JE, Ryan L, Hoover HC, et al. (2000) Adjuvant active specific immunotherapy for stage II and III colon cancer with an autologous tumor cell vaccine: Eastern Cooperative Oncology Group Study E5283. J Clin Onc 18: 148-157. |
[126] |
129. Uyl-de Groot CA, Vermorken JB, Hanna MG, et al. (2005) Immunotherapy with autologous tumor cell-BCG vaccine in patients with colon cancer: a prospective study of medical and economic benefits. Vaccine 23: 2379-2387. doi: 10.1016/j.vaccine.2005.01.015
![]() |
[127] |
130. Schulze T, Kemmner W, Weitz J, et al. (2009) Efficiency of adjuvant active specific immunization with Newcastle disease virus modified tumor cells in colorectal cancer patients following resection of liver metastases: results of a prospective randomized trial. Cancer Immunol Immunother 58: 61-69. doi: 10.1007/s00262-008-0526-1
![]() |
[128] |
131. Parkhurst MR, Yang JC, Langan RC, et al. (2011) T cells targeting carcinoembryonic antigen can mediate regression of metastatic colorectal cancer but induce severe transient colitis. Mol Ther 19: 620-626. doi: 10.1038/mt.2010.272
![]() |
[129] |
132. Chung KY, Gore I, Fong L, et al. (2010) Phase II study of the anti-cytotoxic T-lymphocyte-associated antigen 4 monoclonal antibody, tremelimumab, in patients with refractory metastatic colorectal cancer. J Clin Onc 28: 3485-3490. doi: 10.1200/JCO.2010.28.3994
![]() |
[130] |
133. Brahmer JR, Drake CG, Wollner I, et al. (2010) Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Onc 28: 3167-3175. doi: 10.1200/JCO.2009.26.7609
![]() |
[131] |
134. Lipson EJ, Sharfman WH, Drake CG, et al. (2013) Durable cancer regression off-treatment and effective reinduction therapy with an anti-PD-1 antibody. Clin Cancer Res 19: 462-468. doi: 10.1158/1078-0432.CCR-12-2625
![]() |
[132] | 135. Dela Cruz CS, Tanoue LT, Matthay RA (2011) Lung Cancer: Epidemiology, Etiology, and Prevention. Clin Chest Med 32. |
[133] |
136. Quoix E, Sequist L, Nemunaitis J, et al. (2014) TG4010 immunotherapy combined with first-line therapy in advanced non-small cell lung cancer (NSCLC): phase IIb results of the TIME study. J Immunother Cancer 2: O12. doi: 10.1186/2051-1426-2-S3-O12
![]() |
[134] |
137. Lynch TJ, Bondarenko I, Luft A, et al. (2012) Ipilimumab in combination with paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-small-cell lung cancer: results from a randomized, double-blind, multicenter phase II study. J Clin Onc 30: 2046-2054. doi: 10.1200/JCO.2011.38.4032
![]() |
[135] |
138. Brahmer JR, Horn L, Gandhi L, et al. (2014) Nivolumab (anti-PD-1, BMS-936558, ONO-4538) in patients (pts) with advanced non-small-cell lung cancer (NSCLC): Survival and clinical activity by subgroup analysis. J Clin Onc 32: 5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[136] |
139. Topalian SL, Hodi FS, Brahmer JR, et al. (2012) Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 366: 2443-2454. doi: 10.1056/NEJMoa1200690
![]() |
[137] | 140. Garon EB, Gandhi L, Rizvi N, et al. (2014) Lba43antitumor Activity of Pembrolizumab (pembro|Mk-3475) and Correlation with Programmed Death Ligand 1 (pd-L1) Expression in a Pooled Analysis of Patients (pts) with Advanced Non–Small Cell Lung Carcinoma (nsclc). Ann Onc 25: mdu438. |
[138] |
141. Johnson DB, Rioth MJ, Horn L (2014) Immune Checkpoint Inhibitors in NSCLC. Curr Treat Options Onc 15: 658-669. doi: 10.1007/s11864-014-0305-5
![]() |
[139] | 142. Spigel DR, Gettinger SN, Horn L, et al. (2013) Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic non-small cell lung cancer (NSCLC). J Clin Onc 31. |
[140] |
143. Brahmer JR, Rizvi NA, Lutzky J, et al. (2014) Clinical activity and biomarkers of MEDI4736, an anti-PD-L1 antibody, in patients with NSCLC. J Clin Onc 32:5s. doi: 10.1200/JCO.2013.49.4757
![]() |
[141] | 144. Antonia SJ, Gettinger SN, Chow LQM, et al. (2014) Nivolumab (anti-PD-1|BMS-936558, ONO-4538) and ipilimumab in first-line NSCLC: Interim phase I results. J Clin Onc 32:5s. |
1. | Chad Abresch, Carol Gilbert, Marilyn Johnson, Bibhusha Karki, Kiara Lyons, Karly Meyer, Melissa Tibbits, Drissa Toure, Understanding the Emotional Labor of Public Health Equity Work: a Mixed Methods Study, 2022, 2197-3792, 10.1007/s40615-022-01292-9 | |
2. | Zohra Saleem, Ayaz Muhammad Hanif, Zhou Shenbei, A Moderated Mediation Model of Emotion Regulation and Work-to-Family Interaction: Examining the Role of Emotional Labor for University Teachers in Pakistan, 2022, 12, 2158-2440, 215824402210995, 10.1177/21582440221099520 | |
3. | Lei Pan, Chongyao Yan, Yuan Zheng, Qiang Fu, Yangjie Zhang, Zhiwei Lu, Zhiqing Zhao, Jun Tian, Fatigue detection method for UAV remote pilot based on multi feature fusion, 2022, 31, 2688-1594, 442, 10.3934/era.2023022 | |
4. | Mengru Bu, Haiqi Ma, Huimin Zhai, Yue Ma, Ningjun Xu, Role of self‐efficacy in nursing organizational climate: A way to develop nurses' humanistic practice ability, 2022, 30, 0966-0429, 2107, 10.1111/jonm.13516 | |
5. | Julie Christine Krueger, 2022, chapter 13, 9781668441282, 230, 10.4018/978-1-6684-4128-2.ch013 | |
6. | O. О. Sydorenko, PSYCHOCORRECTION OF PSYCHOLOGICAL MALADAPTATION AMONG CIVIL SERVANTS, 2022, 1, 2616-4868, 59, 10.31612/2616-4868.1(19).2022.08 | |
7. | Yi Sima, Jizheng Yi, Aibin Chen, Ze Jin, Automatic expression recognition of face image sequence based on key-frame generation and differential emotion feature, 2021, 113, 15684946, 108029, 10.1016/j.asoc.2021.108029 | |
8. | Da-Jung Ha, Jung-Hyun Park, Su-Eun Jung, Boram Lee, Myo-Sung Kim, Kyo-Lin Sim, Yung-Hyun Choi, Chan-Young Kwon, The Experience of Emotional Labor and Its Related Factors among Nurses in General Hospital Settings in Republic of Korea: A Systematic Review and Meta-Analysis, 2021, 13, 2071-1050, 11634, 10.3390/su132111634 | |
9. | Ángeles Pastor-López, Pedro E. Ventura-Puertos, José Hernández-Ascanio, Vanesa Cantón-Habas, Pablo Martínez-Angulo, Manuel Rich-Ruiz, Emotional Universe of Nurse Case Managers Regarding Care for Elderly at Risk in Spain: A Hermeneutical Study, 2022, 19, 1660-4601, 16445, 10.3390/ijerph192416445 | |
10. | Ewa Giermanowska, Mariola Racław, Dorota Szawarska, 2023, 9788323581161, 10.31338/uw.9788323581161 | |
11. | Mi-Na Kim, Yang-Sook Yoo, Ok-Hee Cho, Kyung-Hye Hwang, Emotional Labor and Burnout of Public Health Nurses during the COVID-19 Pandemic: Mediating Effects of Perceived Health Status and Perceived Organizational Support, 2022, 19, 1660-4601, 549, 10.3390/ijerph19010549 | |
12. | Fengping Han, Aihua Li, Dongmei Zhang, Lanting Lv, Qian Li, Jing Sun, Petri Böckerman, Relationship between emotional labor and sense of career success among community nurses in China, Beijing: A cross-sectional study based on latent class analysis, 2022, 17, 1932-6203, e0268188, 10.1371/journal.pone.0268188 | |
13. | Duri Lee, Kyungmin Nam, Uichin Lee, 2024, Supporting Interpersonal Emotion Regulation of Call Center Workers via Customer Voice Modulation, 9798400703317, 1, 10.1145/3613905.3650968 | |
14. | Seong-Uk Baek, Jin-Ha Yoon, Jong-Uk Won, Association between high emotional demand at work, burnout symptoms, and sleep disturbance among Korean workers: a cross-sectional mediation analysis, 2023, 13, 2045-2322, 10.1038/s41598-023-43451-w | |
15. | Constantine Sedikides, Rebecca J. Schlegel, Distilling the concept of authenticity, 2024, 3, 2731-0574, 509, 10.1038/s44159-024-00323-y | |
16. | Qiang Yu, Chongmei Huang, Jin Yan, Liqing Yue, Yusheng Tian, Jiaxin Yang, Xuting Li, Yamin Li, Yuelan Qin, Ethical climate, moral resilience, and ethical competence of head nurses, 2024, 0969-7330, 10.1177/09697330241230526 | |
17. | Carol Gilbert, Marilyn Johnson, Bibhusha Karki, Kiara Lyons, Melissa Tibbits, Drissa Toure, Aislinn C. Rookwood, Chad Abresch, Preventing Job Burnout: Could Workplace Support Protect Maternal and Child Health Professionals Who Are Doing Public Health Equity Work?, 2024, 28, 1092-7875, 24, 10.1007/s10995-023-03846-7 | |
18. | Heidi Marie Meling, Sabine Ruths, Valborg Baste, Gunnel Hensing, Inger Haukenes, Level of education and sustainable return to work among long-term sick-listed workers with depression: a register-based cohort study (The Norwegian GP-DEP Study), 2023, 13, 2044-6055, e072051, 10.1136/bmjopen-2023-072051 | |
19. | Hsiu‐Chuan Wu, Chi‐Ho Hsu, Kuan‐Yang Chen, Relationship between nurses’ mood at work and work engagement: Work morale as a mediator, 2024, 71, 0020-8132, 602, 10.1111/inr.12886 | |
20. | Dan Weijers, Nick Munn, 2024, Chapter 10, 978-3-031-59143-3, 169, 10.1007/978-3-031-59144-0_10 | |
21. | Ejura Yetunde Salihu, Promise Tewogbola, Deborah Tolani Joseph, Joy Ofuokwu-Oduniyi, Betty Chewning, Compassion fatigue and satisfaction among pharmacists: Prevalence and associated factors, 2025, 15517411, 10.1016/j.sapharm.2025.04.003 | |
22. | Shohei Okura, Shinichi Iwasaki, Yasuhiko Deguchi, Yuki Kageyama, Kunio Maekubo, Ayaka Matsunaga, Koki Inoue, The associations between workplace gender composition and occupational stress among Japanese public servants, 2025, 1051-9815, 10.1177/10519815251332854 | |
23. | Luis Felipe Dias Lopes, Deoclécio Junior Cardoso Da Silva, Mauren Pimentel Lima, Beatriz Leite Gustmann Castro, Nuvea Kuhn, Fillipe Grando Lopes, Eduarda Grando Lopes, Luciano Amaral, Saúde emocional no local de trabalho: validação de um inventário de medição, 2025, 42, 2145-4515, 10.12804/revistas.urosario.edu.co/apl/a.14070 | |
24. | Tímea Zsuzsanna Popucza, Lars Eriksson, Mårten Eriksson, Emotional demands, burnout, and mental wellbeing in healthcare, care, and service work: the mediating role of surface acting across age, 2025, 3, 2813-771X, 10.3389/forgp.2025.1628713 |
% (N) or mean (SD) | |
Age | |
45-64 | 73.2 (229) |
65+ | 26.8 (84) |
Education | |
< High school | 18.2 (57) |
High school | 23.0 (72) |
Some college | 31.0 (97) |
College + | 27.8 (87) |
Income | |
< $25,000 | 45.7 (143) |
$25,000-$49, 999 | 30.7 (96) |
$50,000-$74, 999 | 13.4 (42) |
$75,000 or more | 10.2 (32) |
No health insurance | 16.0 (50) |
No Usual provider | 6.4 (20) |
Self-reported health | |
Poor | 7.4 (23) |
Fair | 24.3 (76) |
Good | 43.7 (137) |
Very good | 20.4 (64) |
Excellent | 4.2 (13) |
Negative experience with health provider | 43.8 (176) |
Trust measures |
|
Fidelity | 3.82 (1.004) |
Competence | 3.86 (0.986) |
Honesty | 3.96 (0.993) |
Confidentiality | 4.07 (1.012) |
Global trust | 3.85 (1.041) |
Trust index score |
3.91 (1.007) |
*Scores are based on a scale of 1 to 5, with higher scores indicating higher trust. †Five trust measures were summed and averaged. |
% (N) or mean (SD) | |
Age | |
45-64 | 73.2 (229) |
65+ | 26.8 (84) |
Education | |
< High school | 18.2 (57) |
High school | 23.0 (72) |
Some college | 31.0 (97) |
College + | 27.8 (87) |
Income | |
< $25,000 | 45.7 (143) |
$25,000-$49, 999 | 30.7 (96) |
$50,000-$74, 999 | 13.4 (42) |
$75,000 or more | 10.2 (32) |
No health insurance | 16.0 (50) |
No Usual provider | 6.4 (20) |
Self-reported health | |
Poor | 7.4 (23) |
Fair | 24.3 (76) |
Good | 43.7 (137) |
Very good | 20.4 (64) |
Excellent | 4.2 (13) |
Negative experience with health provider | 43.8 (176) |
Trust measures |
|
Fidelity | 3.82 (1.004) |
Competence | 3.86 (0.986) |
Honesty | 3.96 (0.993) |
Confidentiality | 4.07 (1.012) |
Global trust | 3.85 (1.041) |
Trust index score |
3.91 (1.007) |
*Scores are based on a scale of 1 to 5, with higher scores indicating higher trust. †Five trust measures were summed and averaged. |