INNV-22. FACTORS GUIDING THE INITIATION OF TUMOR TREATING FIELDS (TTFIELDS; 200 KHZ) THERAPY FOR GLIOBLASTOMA: SELF-REPORTED PATIENT AND ONCOLOGIST PERSPECTIVES

2021 ◽  
Vol 23 (Supplement_6) ◽  
pp. vi109-vi110
Author(s):  
Peggy Frongillo ◽  
P Gage Gwyn ◽  
Connie Wagenknecht ◽  
Nichelle Renae Adams

Abstract BACKGROUND The standard-of-care for newly-diagnosed glioblastoma (ndGBM) has been the standard Stupp protocol. In ndGBM, approved TTFields (200 KHz) concomitant with maintenance temozolomide significantly improved progression-free survival and overall survival. TTFields-therapy selectively disrupts cancer cell division, requiring array-application to the shaved-scalp to non-invasively deliver TTFields to tumor location and confer clinical benefit. This survey-study assessed factors impacting decision of oncologist/patient/caregiver to initiate TTFields-therapy. METHODS A clinical-market research group administered double-blinded, online-questionnaires (30-min) to oncologists/ndGBM-patients/caregivers. Survey questions, an amalgamate of closed-/open-ended questions, were designed to track awareness, perceptions, and acceptance of ndGBM treatments in a representative United States (US) oncologist and adult patient/caregiver population (semi-annually; 2018-2020). The present survey-analysis focuses on results related to initiation of TTFields-therapy for ndGBM. RESULTS Four separate patient/caregiver surveys (Q1/2018-Q3/2020; (n=50-51/wave) and 4 separate oncologist surveys (Q2/2019-Q4/2020; n=130/wave) were conducted. Results suggest majority of patients with ndGBM research treatment options, including TTFields-therapy, immediately after surgery and before radiation/initial temozolomide; and initiate TTFields-therapy after radiation/initial temozolomide (ie, before maintenance temozolomide). Patients-reported initially learning about TTFields post-surgery via oncologists, nurses, support groups, social-media, and TTFields web-based resources. Among 97% of current/previous TTFields-users, the key patient-driver for selecting TTFields, mirroring oncologist-driver to recommend, was efficacy/survival benefit. Other highly-rated selection-drivers were anti-mitotic mechanism (93%), improved long-term 5-year survival (87%), sustained quality-of-life (87%), and no travel requirement to start (87%). In latest study wave, reported patient usage-barrier was the inability to subsequently enroll in clinical trials (64%), while the top-reported oncologist prescribing-barrier was concern with patient treatment usage (60%). Also, survey suggests oncologists discuss TTFields with most patients with ndGBM, citing efficacy and National Comprehensive Cancer Network® inclusion as key treatment-initiation drivers. CONCLUSIONS Overall, survey data suggests awareness (100%, aided; 61%-68%, unaided) by US oncologists/patients/caregivers of TTFields (200 kHz) as a viable treatment option for adults with ndGBM.

2021 ◽  
Vol 1 (3) ◽  
Author(s):  
Chantelle C. Lachance ◽  
Khai Tran ◽  
Elizabeth Carson ◽  
Joanne Kim ◽  
David Palma ◽  
...  

Oligometastatic cancer (cancer with a limited number of metastases) represents an intermediate state between cancer confined to a single location in the body and cancer that has metastasized — or spread — widely. One treatment option, for which there is growing interest, is stereotactic ablative radiotherapy, also known as SABR. SABR precisely delivers a high dose of radiation to ablate tumours at specific sites while minimizing the radiation dose to surrounding normal tissues. SABR may be used independently or alongside other treatment options in the management of oligometastatic cancer. This CADTH clinical review evaluated the evidence regarding the clinical effectiveness and safety of SABR with or without standard of care (SOC) for people with oligometastatic cancer and found the following: SABR in addition to SOC may offer a benefit in terms of overall survival (OS) and progression-free survival (PFS). The findings for the effectiveness of SABR alone compared with SOC were mixed and deemed inconclusive. There are insufficient data related to adverse events (AEs) at the present time to draw conclusions regarding the safety of SABR relative to SOC alternatives. Note that the CADTH Clinical Review Report will be updated every 3 months to ensure the findings remain up-to-date as new evidence emerges.


2021 ◽  
Author(s):  
Francesca Spada ◽  
Davide Campana ◽  
Giuseppe Lamberti ◽  
Riccardo Laudicella ◽  
Renato Dellamano ◽  
...  

Abstract Purpose To assess and compare clinical outcomes and costs, to the Italian healthcare system, of three therapeutic options approved in the management of adult patients with gastro-enteropancreatic neuroendocrine tumours (GEP-NETs). Methods We compared the efficacy, safety and costs of [177Lu]Lu-DOTA-TATE, everolimus (both originator and generic products) and sunitinib in patients with advanced GEP-NETs (NET G1 and G2) that had progressed following treatment with somatostatin analogues (SSAs). A cost-consequence model was developed and validated by a panel of clinical experts from three NET reference centres in Italy. The clinical outcomes included in the model were median progression-free survival and the incidence of grade 3 or 4 adverse events (AEs), as reported in pivotal clinical trials. The costs for acquisition and administration of each treatment, and of managing AEs, were calculated from the perspective of the Italian national health service. Treatment costs per progression-free month were calculated separately for patients with NETs of pancreatic (PanNETs; all three treatments) and gastrointestinal (GI-NETs; [177Lu]Lu-DOTA-TATE and everolimus only) origin. Results In patients with PanNETs, total costs per progression-free month were €2989 for [177Lu]Lu-DOTA-TATE, €4975 for originator everolimus, €3472 for generic everolimus, and €5337 for sunitinib. In patients with GI-NETs, total costs per progression-free month were €3189 for [177Lu]Lu-DOTA-TATE, €4990 for originator everolimus, and €3483 for generic everolimus. Conclusions [177Lu]Lu-DOTA-TATE was associated with lower costs per progression-free month versus relevant treatment options in patients with GI-NETs or PanNETs (NET G1–G2; progressed following SSA treatment), although acquisition and administration costs are higher. These findings provide further economic arguments in the overall context of treatment decision making.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e16081-e16081 ◽  
Author(s):  
Camillo Porta ◽  
Vittorio D. Ferrari ◽  
Paolo Andrea Zucali ◽  
Giuseppe Fornarini ◽  
Antonio Bernardo ◽  
...  

e16081 Background: Sunitinib is a 1st-line standard of care in mRCC. Lack of cross-resistance to sequential VEGF-targeting drugs and the possibility of a successful rechallenge with Sunitinib have been postulated. Whether mRCC patients (pts) could benefit from rechallenge with Sunitinib after progressing on 1st-line Sunitinib and 2nd-line Everolimus was the aim of this phase II study Methods: 39 mRCCpts were prospectively treated with Sunitinib (50 mg/daily, 4:2); main inclusion criteria were: histologically proven RCC with clear cell component, previous 1st-line Sunitinib with a Disease Control Rate lasting at least 10 months, 2nd-line Everolimus, and written informed consent. The primary end-point of this study was 6-months progression-free survival (PFS). A Simon’s 2-stage design was used; after testing Sunitinib on 12 pts in the first stage, the trial would have been terminated if 5 or fewer had a PFS of less than 6 months. Otherwise, the trial would have proceeded to the second stage, enrolling a total of 38 pts. If the total number of pts free of progression at 6 months would have been less than or equal to 18, Sunitinib would have been rejected Results: As a whole, 39 pts (30 males, 9 females) were enrolled. The study quickly moved from the first stage to its completion and ultimately succeeded; indeed, 6-months PFS was 60%, median PFS being 8.6 months (average: 9.59, range: 0.7-24.6 months). In terms of safety no unexpected toxicities were observed. Tx-related grade 3-4 AEs observed in ≥5% of the pts were: hand-foot skin reaction, fatigue, nausea, hypertriglyceridemia, hypophosphatemia, hypocalcemia, hyperglycemia, and neutropenia. One case each of myocardial infarction, atrial flutter and spontaneous pneumothorax were also reported, but resolved Conclusions: Despite an ineluctable time-lead-bias, median PFS on Sunitinib rechallenge was high (8.6 months), clearly showing that many pts may become sensitive again to VEGFRs-inhibition. Although many agents are presently available from 2nd-line on, in countries where treatment options are still limited, Sunitinib rechallenge could still represent a reasonable treatment option. EudraCT number: 2012-000473-23. Clinical trial information: 2012-000473-23.


2012 ◽  
Vol 30 (4_suppl) ◽  
pp. 607-607 ◽  
Author(s):  
Mary E. Charlton ◽  
Chi Lin ◽  
Jane F Pendergast ◽  
Elizabeth A. Chrischilles ◽  
Robert B Wallace

607 Background: Adjuvant or neoadjuvant chemotherapy (chemo) and radiation therapy (RT) have been the standard of care for virtually all patients with stages II/III rectal cancer for 20 years. However, recent analyses of the SEER database have demonstrated variation in treatment. Leveraging the clinical characteristics and patient beliefs available in the Cancer Care Outcomes Research and Surveillance Consortium (CanCORS) cohort, we determined the proportion and associated characteristics of those who received chemo and RT. Methods: We identified CanCORS patients with stage II/III rectal cancer with primary site resection, and used data from patient surveys and abstracted medical records to construct variables (age, gender, marital status, race, education, income, insurance status, stage, comorbidity, patient treatment beliefs, staging procedures, tumor location/size, types of providers seen). Results: Of the 310 eligible patients, 14% received neither chemo nor RT, 13% received chemo or RT, and 73% received both. Receipt of chemo and RT was associated with younger age, stage III, rectosigmoid junction (vs. rectum NOS) tumor, receipt of staging PET, and patient beliefs that chemo/RT would help them live longer, cure their cancer and help with symptoms. In a logistic model, age (<55 vs >65; OR=2.2, 95% CI: 1.31, 3.46), stage (III vs. II; OR=1.52, 95% CI: 1.06, 2.17), PET (OR=2.41, 95% CI: 1.15, 5.03) and rectosigmoid junction tumor (OR=1.59, 95% CI: 1.10, 2.29) were associated with receipt of chemo; similarly, age and rectosigmoid junction tumor were associated with RT. Of those who did not receive chemo or RT, 35% reported they did not think chemo/RT would help, 65% reported their doctor said chemo/RT would not help and 18% were worried about side effects; 65% reported they never saw a medical or radiation oncologist and 26% reported that no doctor ever discussed the possibility of chemo or RT. Conclusions: A significant proportion of patients with stage II/III rectal cancer did not receive chemo and RT, and a number of those reported that no doctor ever discussed the possibility of chemo or RT with them. While deviation from guidelines may be warranted and/or patient driven, further analyses should determine reasons for lack of recommended treatments.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. TPS2580-TPS2580
Author(s):  
Wenyin Shi ◽  
Lawrence Kleinberg ◽  
Suriya A. Jeyapalan ◽  
Samuel Aaron Goldlust ◽  
Seema Nagpal ◽  
...  

TPS2580 Background: Tumor treating fields (TTFields) is a non-invasive, regional antimitotic treatment approved as a standard of care for glioblastoma (GBM). In the EF-14 phase III trial, TTFields (200 kHz) plus temozolomide (TMZ) significantly increased the survival of patients with newly diagnosed GBM (ndGBM) without increase in systemic toxicity. TTFields-related AEs were mainly skin AEs. In preclinical models, TTFields increase the therapeutic effects of radiation therapy (RT). A pilot study showed that TTFields concomitant with RT and TMZ is well tolerated. The benefit of concomitant TTFields with RT and TMZ will be tested in this phase III TRIDENT randomized trial. Methods: TRIDENT is an international phase III randomized trial comparing standard RT with TMZ vs the triple combination of RT plus TMZ with concomitant TTFields. RT is delivered through the TTFields arrays. Patients in both arms will receive maintenance TTFields with TMZ. TTFields (200 KHz) will be delivered >18 hours/day using Optune. Patients will continue TTFields treatment until second recurrence. Patients with pathologically confirmed newly ndGBM, ≥ 18 years, KPS ≥ 70, either sex, post-surgery or biopsy, who are amenable for RT/TMZ therapy will be enrolled. Patients will be stratified by extent of resection and MGMT promoter methylation status. The primary endpoint is overall survival (OS). Secondary end points include: progression free survival (PFS; RANO), 1- and 2-year survival rates, overall radiological response (ORR; RANO), progression-free survival (PFS2, PFS6, PFS12); severity and frequency of AEs (CTCAE V5.0); pathological changes in resected GBM tumors post treatment; quality of life (EORTC QLQ-C30); and correlation of OS to TTFields compliance. The hypothesis is that concomitant TTFields with radiation and TMZ will significantly improve OS as compared to radiation and TMZ alone. The sample size is 950, with 475 in each arm to detect a HR <0.8 with a 5% type I error. Survival will be measured from the time of randomization until date of death. At the time of analysis, patients who are lost to follow-up or still on protocol follow-up will be censored at the last date known to be alive.


2020 ◽  
Vol 2 (Supplement_3) ◽  
pp. ii3-ii3
Author(s):  
Mitsutoshi Nakada

Abstract Background: Tumor treating fields (TTFields) is a non-invasive, regional antimitotic treatment approved as a standard-of-care for glioblastoma. In the EF-14 Phase 3 trial, TTFields (200 kHz) plus temozolomide (TMZ) significantly increased survival of patients with newly diagnosed GBM(ndGBM) without increasing systemic toxicity. TTFields-related AEs were mainly skin AEs. In preclinical models, TTFields increase the therapeutic effects of radiation therapy (RT). A pilot study showed that TTFields concomitant with RT and TMZ is well tolerated. The benefit of concomitant TTFields with RT and TMZ will be tested in the TRIDENT trial. Methods: TRIDENT is an international phase III randomized trial comparing standard RT/TMZ vs the triple combination of RT/TMZ with concomitant TTFields. RT is delivered through the TTFields arrays. Patients in both arms will receive maintenance TTFields/TMZ. TTFields (200 kHz) will be delivered over18 hours/day using Optune. Patients will continue TTFields treatment until second recurrence. Patients with pathologically confirmed ndGBM, over 18 years old, KPS over 70, either sex, post-surgery or biopsy, and amenable for RT/TMZ therapy will be stratified by extent of resection and MGMT promoter methylation status. The primary endpoint is overall survival (OS). Secondary end points: progression free survival (PFS; RANO), 1- and 2-year survival rates, overall radiological response (ORR; RANO), progression-free survival (PFS2, PFS6, PFS12); severity and frequency of AEs (CTCAE V5.0); pathological changes in resected GBM tumors post treatment; quality of life (EORTC QLQ-C30); and correlation of OS to TTFields compliance. The hypothesis is that concomitant TTFields/RT/TMZ will significantly improve OS versus RT/TMZ. Sample size (N=950; 475/arm) will detect a HR lower than 0.8 with 5% type I error. Survival will be measured from the time of randomization until date of death. At the time of analysis, patients lost to follow-up or still on protocol follow-up will be censored at the last date known to be alive.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. e15624-e15624
Author(s):  
Dadasaheb B Akolkar ◽  
Timothy Crook ◽  
Darshana Patil ◽  
Anantbhushan Ranade ◽  
Amit Bhatt ◽  
...  

e15624 Background: Treatment of advanced refractory cancers face challenges in non-availability of systemic therapy regimens with evidenced benefit. Post failure of two to three lines of systemic treatments, patients with such cancers are usually considered for palliation or clinical trials. Prior attempts at label-agnostic treatment regimens (precision medicine) in such populations were largely based on a single-indication-single-drug paradigm which had limited application. We hypothesized that advanced refractory malignancies have latent vulnerabilities which can be identified by an integrational multi-analyte interrogation of the tumor, and can be targeted using patient-specific combination regimens to yield clinical benefit. Methods: Fresh tumor tissue and blood samples were obtained from 158 patients with solid organ cancers where the disease had progressed following failure of at least two lines of standard of care systemic treatment options. These samples were used for Encyclopedic Tumor Analysis (ETA) which interrogated gene mutations, gene overexpression, pathway dysregulation, immunohistochemistry as well as in vitro chemosensitivity profiling of viable tumor cells. Integration of datasets from the multi-analyte ETA was used to generate patient-specific therapy recommendations. Patients who received ETA-guided treatments were followed up and response to treatment was retrospectively evaluated from radiological scans. Results: All patients received ETA-guided individualized treatments which were combinations of cytotoxic, targeted and endocrine agents. No two patients received the same treatment regimen. Complete or Partial Response (CR or PR) was observed in 76 patients yielding an Objective Response Rate (ORR) of 48.1%. 67 patients showed Stable Disease (SD), thus yielding a Disease Control Rate (DCR) of 90.5%. Median Progression Free Survival (PFS) was 117 days (Range 27 – 379 days). There were no Grade IV therapy related Adverse Events or therapy related deaths. Conclusions: Viable efficacious combination treatment options can be made available for patients with advanced refractory malignancies via ETA, despite perceived non-availability or non-viability of standard of care treatment options.


2021 ◽  
Vol 11 (6) ◽  
Author(s):  
Sundar Jagannath ◽  
Yi Lin ◽  
Hartmut Goldschmidt ◽  
Donna Reece ◽  
Ajay Nooka ◽  
...  

AbstractPatients with relapsed and refractory multiple myeloma (RRMM) who are triple-class exposed (to an immunomodulatory agent, proteasome inhibitor, and anti-CD38 antibody) have limited treatment options and there is no standard of care. Idecabtagene vicleucel (ide-cel, bb2121), a BCMA-directed CAR T-cell therapy, demonstrated efficacy in triple-class exposed RRMM patients in the KarMMa trial (NCT03361748). In this retrospective study (KarMMa-RW), patient-level data from triple-class exposed RRMM patients were merged into a single data model and compared with KarMMa using trimmed stabilized inverse probability of treatment weighting. Endpoints included overall response rate (ORR; primary), rate of very good partial response or better (≥VGPR), progression-free survival (PFS), and overall survival (OS). Of 1949 real-world triple-class exposed RRMM patients, 190 received subsequent (index) line of therapy and met KarMMa eligibility criteria (Eligible RRMM cohort). With a median follow-up of 13.3 months in KarMMa and 10.2 months in Eligible RRMM, ORR, and ≥VGPR were significantly improved in KarMMa versus Eligible RRMM (ORR, 76.4% vs 32.2%; ≥VGPR, 57.9% vs 13.7%; both P < 0.0001) as were PFS (11.6 vs 3.5 months; P = 0.0004) and OS (20.2 vs 14.7 months; P = 0.0006). This study demonstrated that ide-cel significantly improved responses and survival compared with currently available therapies in triple-class exposed RRMM.


2017 ◽  
Vol 35 (4_suppl) ◽  
pp. 338-338
Author(s):  
Mark Ricigliano ◽  
William H. Isacoff ◽  
Allyson J. Ocean ◽  
Brandon Cooper ◽  
Mary Welkie ◽  
...  

338 Background: Standard of care treatment for pancreatic adenocarcinoma (PDAC) includes FOLFIRINOX or gemcitabine with nab-paclitaxel used in either the front-line or 2nd line treatment setting. There is no consensus for 3rd line treatment for this cohort of poor prognosis patients with clinical trials being the only viable option. We evaluated a cell viability assay using cultures of circulating tumor and invasive cells (CTICs) to identify effective 3rdline PDAC treatment options. Methods: CTICs were isolated from peripheral blood samples of 17 PDAC patients who previously had received one of the two standard of care regimens using an immunomagnetic separation for EPCAM epitope Ber-EP4 and cultured in enriched DMEM-F12 medium. After 14 days, seven chemotherapeutics (gemcitabine (G), oxaliplatin (O), fluorouracil (F), irinotecan (I), mitomycin C (M), cisplatin (C) and paclitaxel (P)) were added to the cultures and response to treatment was measured 48 hr. after treatment by immunofluorescence using a live-cell resazurin assay. Response to treatment was determined by a reduction of fluorescence in the treated cultures compared to control and the mean florescence (Fm) was calculated for each chemotherapeutic. A one-sample t-test was used to measure the variance between Fm control and treated samples. (p-value <0.005). Sensitivity to treatment was determined using linear regression based on Fm values and the sensitivity of each chemotherapeutic was interpolated from a standard curve (range 1.0-5.0, sensitive <2, intermediate 2-3, resistant >3). Results: m (Fm=1.6) demonstrated the most effective treatment option with marked resistance to treatment for G, I, P and O. (Fm=3.4, 3.7, 3.4 and 3.7). Intermediate response was observed for F and C (Fm=2.8 and 2.8). Patient treatment response to m will be described. Conclusions: CTICs can be routinely isolated and cultured for cell viability assays. m based regimens with C and F should be considered for 3rd line PDAC patients.


2019 ◽  
Vol 1 (Supplement_2) ◽  
pp. ii44-ii44
Author(s):  
Daisuke Shimada ◽  
Keiichi Kobayashi ◽  
Kuniaki Saito ◽  
Yoshie Matsumoto ◽  
Yoshiaki Shiokawa ◽  
...  

Abstract BACKGROUNDS Current standard of care for glioblastoma, consists of postoperative temozolomide (TMZ) concomitant with radiotherapy, followed by adjuvant TMZ monotherapy. Recently, an international phase 3 trial (EF-14) demonstrated that addition of tumor-treating fields (TTF) to adjuvant TMZ after completion of chemoradiotherapy extended median progression-free survival and overall survival by 2.7 months and 4.8 months, respectively, compared with TMZ alone in patients with newly diagnosed glioblastoma. TTF is now considered as a part of its initial treatment in the guideline in Japan (recommendation grade B). However,distinct from anticancer drugs,little is known or experienced using TTF as a therapeutic device so far, especially in management and handling. METHODS First six patients with newly diagnosed glioblastoma who underwent TTF were analyzed with special interest in medical and social supports to execute TTF at home. RESULTS TTF was first introduced in our institution in May 2016, but no patients were treated because of no coverage by medical insurance until December 2017. We further needed to wait to initiate TTF treatment until January 2019 when the contract to use TTF systems was finally made between the company and institution. Since then six patients were registered in five months. For its introduction to clinical practice,it is essential to establish new in-house environment with medical professions division in the facility including documentations of calculating instruction fees and usage guidance for home care application of TTF. It is also important to initiate providing information of TTF such as timing of visit by specific practitioners and potential medical and psychologic burdens to patients and their families during and after chemoradiotherapy to better understand this new modality leading to the consent acquisition. CONCLUSIONS Introducing TTF into clinical practice should accompany improvement of management in not only medical equipment and documentations but also patient care in hospital and home.


Sign in / Sign up

Export Citation Format

Share Document