Quality of life in patients with malignant melanoma participating in a phase I trial of an autologous tumour-derived vaccine

2002 ◽  
Vol 12 (5) ◽  
pp. 505-511 ◽  
Author(s):  
L. Cohen ◽  
P. A. Parker ◽  
J. Sterner ◽  
C. de Moor
2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS12136-TPS12136
Author(s):  
Ishwaria Mohan Subbiah ◽  
Jaya Sheela Amaram-Davila ◽  
Angelique Wong ◽  
Kaoswi Karina Shih ◽  
Aimee Elizabeth Anderson ◽  
...  

TPS12136 Background: Patients w advanced cancer participating in Phase I trials carry a high symptom burden from cancer and prior therapies. Our prior work shows patients on phase I trials w multiple active symptoms impacting their immediate quality of life with implications on toxicities and clinical outcomes on subsequent therapy. To identify an effective scalable approach to comprehensive symptom management for patients w adv cancer on phase I trials, we leveraged the increased technology use to design a technology-enhanced symptom management and palliative care intervention (TEC). Methods: Patients w adv cancer seen in the phase I clinic will be given the Edmonton Symptom Assessment System (ESAS), a validated patient-reported outcomes (PRO) tool of common cancer symptoms to identify those with a high symptom burden defined as ≥4 out of 10 on >1 ESAS symptom and a Global Distress Score (GDS) of ≥20. The GDS, a validated score of overall symptom intensity derived from the ESAS, is comprised of 6 physical (pain, fatigue, nausea, drowsiness, appetite, shortness of breath) & 2 psychosocial symptoms (depression, anxiety), and overall wellbeing. TEC is an innovative patient-centered care program of strategic vigorous symptom management where standard-of-care clinic visits are complemented by proactive symptom monitoring between clinic visits remotely and through provider-initiated calls. In this pilot randomized study, we will determine the effect sizes of High-Intensity TEC (HI-TEC; q3day remote PRO assessments w preset provider-initiated call bw visits), Low-Intensity TEC (LO-TEC; q5day remote PRO assessments w preset provider-initiated call bw visits), and Standard Palliative Care (no preset provider contact bw visits). Our guiding hypothesis is that a comprehensive, proactive, technology-enhanced symptom management program led by a Palliative Care team can mitigate the high symptom burden of patients with advanced cancers enrolling in phase I trials. The primary objective assesses the effect size of each TEC intervention on the GDS measure of symptom burden prior to C1D1 on phase I trial. Our working hypothesis is that HI-TEC and LO-TEC will be associated with a lower overall symptom burden signifying symptom optimization prior to starting on a phase I trial. Secondary objectives aim to estimate the effect size of TEC on the following: Symptom burden over 12 weeks on a phase I trial using ESAS, quality of life using FACIT-Sp, PRO-CTCAE and patient satisfaction using FAMCARE-P13. clinical outcomes at 6 months including OS, treatment outcomes (interruptions, dose reductions, discontinuation, time on trial) and quality metrics for end-of-life (EOL) (chemotherapy in the last 14 days of life, ICU admit in last 30 days of life, death without hospice or < 3d of hospice). Qualitatively assessment of patients’ + caregivers’ perceptions of receiving TEC-based cancer care. Clinical trial information: NCI-2020-07465.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3114-3114
Author(s):  
Craig C. Hofmeister ◽  
Mindy A Bowers ◽  
Yvonne A Efebera ◽  
Kristina Humphries ◽  
Don M. Benson ◽  
...  

Abstract Abstract 3114 Introduction: Post autologous transplant maintenance therapy for patients with multiple myeloma (MM) is standard of care (McCarthy et al, NEJM, 2012). Vorinostat (SAHA, Zolinza) is a HDAC inhibitor for which preclinical evidence suggests that its combination with bortezomib is synergistic via HDAC-6. Preclinical data suggests that HDAC-I's increase MHC class I and class II expression, rendering tumor cells more susceptible to host innate immune killing. Lenalidomide activates NK cells via PP2A inhibition and induces CD56 expression in CD16+CD56- cells thereby enhancing NK cell-mediated ADCC. Initiating lenalidomide to enhance NK cell activity against tumor cells in the early post transplant period, especially if administered after increased MHC class I expression induced by HDAC-I pretreatment. We hypothesized that the combination of Vorinostat and lenalidomize would be both tolerable and effective post-transplant. Methods: This was a phase I trial for myeloma patients after hematopoietic stem cell transplant (HSCT) following the 3×3 phase I design. Patients were required to have an ANC3 1000/μL, platelet count3 75,000/μL, and a serum creatinine2 1.5x institutional upper limit of normal. Vorinostat was administered starting day +90 after HSCT for days 1–7 and 15–21 starting at 200 mg and escalating to a max of 400 mg combined with lenalidomide 10 mg days 1–21 of a 28-day cycle until progression or clinically significant toxicity. Lenalidomide could be escalated after cycle 1 in 5 mg increments to a maximum of 25 mg. The primary endpoint was maximum tolerated dose (MTD) and dose limiting toxicities were assessed during the first cycle. Correlative endpoints included quality of life assessments with the Brief Pain Inventory (Short Form), The Center for Epidemiologic Studies Depression Scale (CES-D-10), Brief Fatigue Inventory (BFI), Brief Pain Inventory (BPI), and the FACT-G. Peripheral blood flow cytometry for NK cells and Tregs were obtained day 1 of the first cycle of combined therapy (C1D1/3 mos. post transplant), C2D1 (4 mos.), C3D1 (5 mos.), C4D1 (6 mos.), and off study. Results: Sixteen patients were enrolled after HSCT with a median age 58 y.o. (range 41–67) with a median number of prior therapies at enrollment of 2 (range 1–8) and mean ISS stage 1.5 (range 1–3). Twelve patients had only trisomies on CD138-selected FISH, one patient had normal cytogenetics, and three patients had high risk features [complicated karyotype, t(4;14), or abnormal chromosome 1]. Median follow-up has not been reached. Fifteen patients received more than one cycle of therapy. 11/15 (73%) were able to escalate the lenalidomide dose with 4/11 (36%) reaching the maximum lenalidomide dose, and all but 1 of the 11 required lenalidomide dose reduction due to subsequent neutropenia. Of the adverse events possibly, probably, or definitely related to therapy (table 1), the most common toxicities were neutropenia (11.8% of all AEs), fatigue (11.1%), thrombocytopenia (9.7%), diarrhea (7.6%), anemia (6.9%), hypokalemia (6.3%), and rash (4.9%). Infection with a normal ANC occurred only twice and no patient suffered a DVT. One patient had therapy stopped due to toxicities, one due to progressive disease, and one due to megacolon that was present prior to transplant. Four patients improved their transplant response after starting lenalidomide/vorinostat. Preliminary analysis of quality of life from the start of therapy through the first three cycles found no significant change in the BFI, FACT-G, CES-D, or BPI. Conclusions: Lenalidomide/vorinostat post HSCT was well tolerated and 14/16 (87%) of patients remain on protocol therapy. There were no DLTs and patients the final cohort starting at 400 mg vorinostat and 10 mg lenalidomide completed accrual. The median dose of lenalidomide for those patients receiving maintenance therapy 1 year post-transplant is 5 mg daily. Analysis of peripheral blood flow NK cells and Tregs, cumulative dose intensity, and median progression free survival will be presented at the meeting. Disclosures: No relevant conflicts of interest to declare.


2017 ◽  
Vol 27 (1) ◽  
pp. 163-170 ◽  
Author(s):  
Diane A. van der Biessen ◽  
Peer G. van der Helm ◽  
Dennis Klein ◽  
Simone van der Burg ◽  
Ron H. Mathijssen ◽  
...  

2009 ◽  
Vol 27 (15_suppl) ◽  
pp. e20709-e20709
Author(s):  
G. Jung ◽  
D. Knight ◽  
A. Moadel ◽  
K. Desai ◽  
I. Chaudhary ◽  
...  

e20709 Background: Quality of life (QoL) assessment in clinical trials has been gaining more attention. FACT-G surveys have been validated to assess QoL in clinical trials involving oncology patient (Cella DF et al, J Clin Oncol 11:570–579, 1993). However, there is paucity of evaluation of QoL in patients with advanced cancer participating in Phase I clinical trials. Methods: FACT-G surveys were conducted within the context of a Phase I trial to identify a safe dose and potential drug-drug interations of capecitabine and irinotecan combination (Goel, S et al, Invest New Drugs 25:237–245, 2007). The FACT-G survey consists of 28 questions in 5 sections, namely, physical well-being, social/family well-being, emotional well-being, relationship with doctor, and functional well-being). Patients were requested to complete the FACT-G surveys at baseline and every two cycles thereafter (each cycle of 3 weeks duration). Results: Forty-one of 47 patients with advanced solid tumors who participated in the clinical trial completed FACT-G surveys. Mean scores were calculated for each time point. The mean QoL scores at baseline and post cycle 2 were 53 and 58, respectively (p = 0.1). Post cycle 4, the mean QoL score was 62 [p = 0.01, (vs. baseline)]. Following cycle 4, the number of respondents decreased to the extent where we were unable to ascertain any further changes in the QoL scores. Conclusions: It is feasible to use FACT-G survey as a tool to assess QoL in patients participating in an oncology phase I clinical trial. Although the sample size of the patient population was not powered for any statistical significance, there was a trend toward improving QoL based on FACT-G survey scores. This suggests that phase I clinical trials may provide improvement of QoL for some patients. FACT-G is a useful tool in assessing QoL in oncology phase I trial study population. No significant financial relationships to disclose.


2005 ◽  
Vol 23 (16_suppl) ◽  
pp. 8217-8217 ◽  
Author(s):  
B. F. Issell ◽  
C. Gotay ◽  
I. Pagano ◽  
A. Franke

2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 19670-19670
Author(s):  
J. Y. Bruce ◽  
F. J. Hlubocky ◽  
C. K. Daugherty

19670 Background: Subjects who enroll in phase I cancer trials generally are those with advanced and terminal disease. Although all such patients (pts) undergo an informed consent process, there is evidence that these pts may not appreciate the primary aims of a phase I trial and have unrealistic expectations of therapeutic benefit. However, these perceptions of benefit have not been described in pts after trial enrollment. Methods: As a pilot study, pts previously enrolled on Phase I trials were interviewed regarding their retrospective perceptions of benefit. A total of 9 pts have been interviewed to date: median age: 72 yr (range: 59–82 yr); 55% male; 77% Caucasian. The interview included open-ended questions assessing patient experiences with the trial, including reasons for enrollment, impressions of quality of life, costs incurred, and other benefits. Pts’ qualitative responses were recorded and analyzed for content and themes. Results: Reasons for enrollment included hope, prior treatment failure, altruism, and family influence. Described perceptions of anticipated benefit included stabilization of disease and close monitoring of their disease. Unexpected costs involved time spent during clinic visits, financial costs, time taken off work by family members, time spent away from family, and additional trial procedures. Issues of quality of life revolved around side effects from the investigational agent. Many interviewed pts (44%) described improved quality of life off the trial as compared to during trial participation. The majority of interviewed pts (67%) believed that the trial had no effect on their survival. Conclusions: Our data suggest that pts’ perceptions of therapeutic benefit after trial enrollment may be different from pre-enrollment expectations. There would also appear to be potentially significant and otherwise undescribed out-of-pocket expenses incurred by subjects as a result of phase I enrollment. Further research is needed to examine how side effects, unexpected costs, and lack of improved survival contribute to these pts’ perceptions of trial benefit. No significant financial relationships to disclose.


2018 ◽  
Vol 27 (6) ◽  
pp. e12908
Author(s):  
Diane A. van der Biessen ◽  
Wendy H. Oldenmenger ◽  
Peer G. van der Helm ◽  
Dennis Klein ◽  
Esther Oomen-de Hoop ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document