Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
Filter
Type of Medium
Language
Subjects(RVK)
  • 1
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. e23548-e23548
    Abstract: e23548 Background: Rhabdomyosarcomas are the most common soft tissue sarcoma in children, and the prognosis of pediatric ARMS has improved with the use of multi-modality therapies. However, ARMS in adults is rare, and long-term outcomes continue to be poor. This study aimed to evaluate clinical outcomes of an adult ARMS population on different front-line systemic chemotherapies, particularly the vincristine/doxorubicin/ifosfamide (VDI) regimen. Methods: Adult fusion-positive confirmed ARMS patients over the age of 18 years (y) treated at MDACC from 2004 to 2018 were identified in our patient registry. Descriptive clinical statistics including staging, front-line chemotherapy, multimodal therapy usage, and survival analyses were performed. Results: 49 patients were identified, with mean age of 34.9 y (range 18y - 67y), and 53% were male. Most patients were white (53%, 26 pts), and the most common primary tumor site was the parameningeal space (63%; 31 pts). Patients were either intermediate (67%) or high clinical risk (33%). Most patients were IRSG clinical group IIIa (36%), IIIb (20%) or IV (33%) and were classified clinical stage 3 (49%) or 4 (33%). Of all patients at diagnosis, 71% had nodal disease and 32% were metastatic. Radiotherapy and surgery were given with upfront chemotherapy in 33 pts (67%) and 24 pts (49%) respectively, with 19 patients receiving both. Median OS for the entire cohort was 3.6 years. Doxorubicin containing chemotherapy regimens trended to worse OS than non-doxorubicin containing regimens (2.3 yrs vs 4.0 yrs, p = 0.355). Comparing patients who received VDI (19 pts) vs non-VDI (30 pts; 13 received Actinomycin D, 12 received doxorubicin in different regimens, and 5 received neither), median OS was 1.8 yrs vs 3.8 yrs (p = 0.283) respectively. There were similar number of front-line chemotherapy cycles (8.5 vs 9.5 cycles), high clinical risk (26% vs 37%) and metastatic disease (21% vs 36%) in the VDI vs non-VDI cohorts. Patients receiving upfront radiation had improved survival (3.7 vs 1.5 yr, p = 0.01), but this is likely confounded by those with metastases being less likely to receive upfront radiation. Conclusions: In this single center retrospective analysis of adult ARMS patients, survival outcomes continue to be similar to historical outcomes. There was no statistically significant OS difference in patients who did or did not receive doxorubicin containing front-line chemotherapy regimens, or in particular VDI therapy, although there was a trend to decreased OS. However, limitations to this study include limited sample size, non-randomization to treatment selection, and possible biases in patient selection for different chemo regimens. Based on these observations, randomized prospective studies are necessary to delineate which frontline chemotherapy regimen is most beneficial in this rare tumor in adults.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 11549-11549
    Abstract: 11549 Background: Targeting the DNA damage response (DDR)/Homologous Recombination (HR) pathway is an emerging therapeutic approach for leiomyosarcoma (LMS). Loss of RNase H2 decreases DNA repair via the Non-Homologous End-Joining (NHEJ) pathway, leading to increased double stranded breaks, replication stress, and increased cell death. Therefore, we developed an assay to screen for RNase H2 loss in STS patient samples to determine its prevalence and prognostic significance, particularly in LMS patients. Methods: RNASEH2B homozygous deletion (HomDel) calls from TCGA samples were based on the Allele-Specific Copy number Analysis of Tumors (ASCAT2) algorithm. Immunohistochemistry (IHC) of RNase H2 was performed on tissue microarrays (TMAs) of uterine (U-LMS) and soft tissue (ST-LMS) leiomyosarcoma samples from MD Anderson Cancer Center (MDACC) using a selective antibody developed by Repare Therapeutics. RNase H2 loss was defined as 〈 = 10% cells without nuclear IHC staining and scored by experienced pathologists in 2 separate cohorts. Genomic analysis was performed using SNiPDx™, a targeted-NGS assay designed to detect biallelic loss of function in select DDR related genes and whole genome sequencing (WGS). MDACC TMA samples were clinically annotated by retrospective review. Results: Using TCGA data, RNASEH2B HomDels were seen in 6% (5/80) of all LMS cases, with a higher proportion in U-LMS (15%; 4/27) as compared to ST-LMS (2%; 1/53). In a pan-tumor TMA, RNase H2 loss by IHC was found in 3.8% (32/843) of samples overall, and in 10% (9/88) of LMS samples. This proportion of RNase H2 loss was higher in a larger MDACC LMS cohort, where negative staining of RNase H2 was found in 30% (33/110) of U-LMS and 38% (39/102) of ST-LMS cases. 30 MDACC LMS cases (15 U-LMS and 15 ST-LMS) were analyzed for RNASEH2B HomDels by SNiPDx. In U-LMS, RNASEH2B HomDels were detected in 64% (7/11) of RNase H2 IHC loss cases vs. 0% (0/3) in RNase H2 IHC intact cases. No RNASEH2B HomDels were detected in ST-LMS cases (10 IHC loss, 5 intact). The median overall survival (mOS) of MDACC U-LMS patients (n = 109) was 4.3 years. No significant mOS difference was seen in RNase H2 IHC intact cases versus loss (mOS 4.4 v 3.3 years, p = 0.54). In a separate cohort, 45 U-LMS samples were screened by IHC; 22% (10/45) had RNase H2 loss. RNASEH2B HomDels were detected in 70% (7/10) of RNase H2 IHC loss cases using SNiPDx and confirmed using WGS. In contrast, RNASEH2B HomDels were detected in 3% (1/33) of RNase H2 IHC intact cases. In the combined U-LMS cohort with IHC and SNiPDx results (n = 71), the diagnostic accuracy, sensitivity and specificity of RNase H2 IHC for detecting RNASEH2B HomDels was 76%, 93% and 71% respectively. Conclusions: RNase H2 loss via RNASEH 2B HomDels is prevalent in U-LMS and is unique from other DDR pathway alterations. RNase H2 IHC is an effective screening tool and is being developed for future clinical trials targeting DDR in LMS.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 39, No. 28_suppl ( 2021-10-01), p. 280-280
    Abstract: 280 Background: The COVID-19 pandemic has negatively impacted morbidity, mortality, and economic status globally. Adolescents and young adults (AYAs, age 15-39) with cancer may experience disparities in access to health care, compounded by social distancing and stay-at-home orders aimed at reducing the spread of COVID-19. The use of telehealth platforms for clinical visits has accelerated rapidly due to policy changes during the pandemic. Telehealth may provide an avenue for accessing healthcare services among AYAs with cancer; yet, there are few data on AYAs’ preferences and satisfaction relating to telehealth. Our study examined telehealth utilization, satisfaction, preferences and future recommendations among AYAs with a cancer diagnosis. Methods: AYAs in active cancer treatment or in post-treatment survivorship completed an online questionnaire that assessed their experience with telehealth, including: satisfaction with the telehealth visit, likelihood of participating in a future telehealth visit, topics participants would like to discuss via telehealth, and open-ended comments regarding suggestions for improving future telehealth visits. Participants were recruited in two cohorts: a pre-vaccine cohort (September 2020 - January 2021) prior to availability of COVID-19 vaccines in Texas; and, a post-vaccine cohort (April 2021 - May 2021) after vaccine availability. Descriptive statistics were used to summarize preliminary findings. Results: Participants included 273 AYAs with cancer (mean age, 33.8 years old, 26% male, 73% non-Hispanic White, 11% in active treatment, 53% in post-vaccine cohort). Of 71.7% who participated in at least one telehealth visit, 50.9% were somewhat or very satisfied with their visit(s). Topics that all participants preferred to address via telehealth included: stress management (34%); sleep quality (31.7%); diet/nutrition (32.1%); and mental health (29.5%).Topics least preferred for telehealth included fertility counseling (16.7%) and sexual health (13.7%). Open-ended comments from participants highlighted the importance of healthcare providers taking time to listen to their concerns during their visits. Participants suggested that telehealth encounters can be improved by ensuring adequate time for the visit plus good video and audio quality, and using telehealth for follow-up visits rather than initial consultations. Conclusions: About half of AYAs who had participated in a telehealth visit indicated satisfaction with the visit(s). Findings indicated AYAs’ preferences for the types of topics that are most and least preferred to address in telehealth visits, as well as specific recommendations for improving the quality of visits. Consideration of AYAs’ preferences and recommendations in the ongoing implementation of telehealth may help improve patient satisfaction.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2021
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. TPS11594-TPS11594
    Abstract: TPS11594 Background: Anti-PD 1/PD-L1 blockade alone or in combination with anti-CTLA4 have yielded suboptimal results in most sarcoma subtypes. CD73, an ectonucleotidase, catalyzes the rate-limiting step for adenosine production in the extracellular space, which then aids tumors in evading immune recognition and destruction. Oleclumab, a monoclonal antibody (mAb) selectively binds and inhibits the activity of CD73, and preclinical data suggests additive activity with durvalumab, a mAb that blocks PD-L1. We designed a trial combining oleclumab and durvalumab in certain sarcoma subtypes, selected based on modest activity with anti-PDL-1 and intense staining with CD73 in the tumor microenvironment. Methods: This phase 2 study (NCT04668300) is enrolling patients with age ≥18 years with recurrent/metastatic angiosarcoma (cohort 1) or dedifferentiated liposarcoma (DLPS) (cohort 2) and ≥12 years with recurrent/metastatic osteosarcoma (cohort 3), who have received at least one prior systemic therapy but are checkpoint inhibitor naïve and have measurable disease. Each treatment cycle is 28 days with oleclumab administered at 3000 mg i.v. every 2 weeks x 5 doses, and then every 4 weeks and durvalumab administered at 1500 mg i.v every 4 weeks. Tumor assessments are based on RECIST 1.1 and immune-related response criteria (irRC) and performed at baseline, and every 8 weeks after start of therapy, with an additional scan at 12 weeks for confirmation of response. Planned sample size is ≤ 25 pts in each arm. The primary efficacy endpoint for cohorts 1 and 2 is response rate (RR) at 4 months (per RECIST 1.1). The primary efficacy endpoint for cohort 3 is event free survival (EFS) rate at 4 months. If there is a high probability that the RR4 months is unlikely to be at least 20% for cohorts 1 and 2 or the EFS4 months is unlikely to be at least 40% for cohort 3 then the accrual of the corresponding cohort will be halted. The cohorts will be monitored separately for both futility and toxicity in groups of 5 after a minimum of 10 patients have been enrolled in each cohort. Secondary endpoints for the study include safety, best RR by RECIST and irRC, median PFS, and OS. Core needle biopsies and blood samples are collected at baseline and early on-treatment (week 6). Fresh flow cytometry is being performed to assess changes in T-cell activation, proliferation, and function and CD73 expression in the membrane and cytoplasm is being assessed by immunohistochemistry. Localization of tumor-infiltrating lymphocytes and engagement of the PD-1/PDL1 axis is being assessed using multiplex immunofluorescence staining. As of Jan 30th, 2022, twenty-two patients have initiated study treatment, 3 in cohort 1, 12 in cohort 2, and 7 in cohort 3. Clinical trial information: NCT04668300.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 12017-12017
    Abstract: 12017 Background: The 5-year survival rates for adolescents and young adults (AYA) with cancer have steadily improved, yet disparities by race/ethnicity persist. The drivers of these disparities remain unclear. Neighborhood socioeconomic deprivation has been linked to adverse health outcomes independent of the effects of individual-level measures of social determinants of health (SDOH). We evaluated the impact of neighborhood SDOH on overall survival (OS) in a diverse AYA cancer patient population treated in a single academic setting. Methods: This study utilized a multi-ethnic cohort of AYA cancer survivors (N=10,261) diagnosed between ages 15-39 seen at MD Anderson Cancer Center between 2000 and 2016. Patient demographics, zip code at presentation, cancer characteristics, and follow-up were obtained from our institutional tumor registry. Zip codes were linked to Area Deprivation Index (ADI) values, a validated neighborhood-level SDOH measure, with higher ADI representing worse SDOH. Results: The cohort included 6,361 (62%) non-Hispanic White, 1,112 (10.8%) non-Hispanic Black, and 2,133 (20.8%) Hispanic AYA cancer patients, with 655 (6.4%) of other ethnicities or missing information. The mean ADI value was significantly higher (p 〈 0.05) for Black (61.7) and Hispanic (65.3) patients, compared to White (51.2) patients. Analysis of ADI by cancer type showed significant differences, mainly driven by the significantly higher ADI in patients with cervical cancer (62.3) than other cancer types. In contrast, AYA patients diagnosed with breast cancer (53.0) and CNS tumors (52.9) resided in neighborhoods with the lowest ADI values. ADI was a significant predictor of OS overall. In multivariable models that included gender, age at diagnosis, cancer diagnosis, and race/ethnicity, risk of death for AYA cancer patients residing in neighborhoods with ADI in the highest quartile was greater than that for patients in the lowest quartile (HR: 1.09, 95% CI: 1.00-1.19, p=0.043). Conclusions: Our results in this large multi-ethnic cohort indicate that non-White AYA patients with cancer more commonly live in neighborhoods associated with higher ADI compared to their White counterparts. Even when treated in the same academic setting, AYA cancer patients with higher ADI experienced worse OS across cancers. Although the magnitude of the effect was moderate, the presence of this effect within a tertiary cancer center with existing barriers to care suggests that ADI is a meaningful risk factor impacting survival. Further analysis is warranted to determine the generalizability of these findings to the patient population more broadly. However, the results provide intriguing evidence for potential interventions aimed at supporting AYA cancer patients from disadvantaged backgrounds.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. 11542-11542
    Abstract: 11542 Background: Ripretinib (R) is approved for 4 th line treatment of GIST based on superior PFS and OS compared to placebo in a phase 3 study, with RR of 11.8% and PFS of 6.3 months (mo). In addition to having high potency against PDGFRA D842V, avapritinib (A) showed activity in 4 th or later lines for KIT mutated patients (pts). The RR was 17% and median duration of response was 10.2 mo from a phase 1 study. It is not known if pts receiving R benefit from A after progression or vice-versa. We retrospectively reviewed outcomes of R and A to determine if sequence affects outcomes. Methods: Pts diagnosed with GISTs and treated with both R and A at UTMDACC from Jan 2016 to Dec 2021 were included. Pts were separated into R-A (RA) or A-R sequence (AR) and outcomes were tabulated. Descriptive statistics were used to summarize characteristics and genetic profiles. Differences between RA and AR groups were calculated using Fisher’s exact. Response was evaluated using RECIST. Kaplan-Meier and Log-rank test were used to estimate and compare PFS and OS between groups. Results: Twenty pts were included in the study; 12 in RA and 8 in AR. Median age was 55 years (R:29.7-76.3). Most pts had small bowel primary (11/20, 55%) followed by stomach (4/20, 20%). All baseline characteristics and mutations were equitably distributed between RA and AR. RR of R was 17% and 14% in RA and AR groups, respectively and RR of A was 33% and 29% in RA and AR, respectively (Table). None of the pts with secondary exon 13 mutations responded to R or A. The drug administration sequence did not affect RR (p = 0.7, 0.62 in R, A). The PFS of the second drug administered, was shortened. PFS did not differ based on type of KIT mutation. Median OS from diagnosis in RA and AR groups were 121.8 (58.2-NR) and 171.3 (73.3-NR) mo, respectively and median OS from start of RA and AR was 43.9 (23.0-NR) and NR (11.5-NR) mo, respectively. Neither difference was statistically significant. Conclusions: Both R and A are efficacious in later lines of treatment, with greater benefit from the agent used first. Although, the combined PFS was numerically higher in AR compared to RA (20.5 vs 15.2 mo), the OS was not different. Currently, R is the only drug approved in the 4 th or later lines in KIT-mutated GIST. [Table: see text]
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 16_suppl ( 2022-06-01), p. TPS11584-TPS11584
    Abstract: TPS11584 Background: Osteosarcoma (OS) is the most common primary bone malignancy of childhood and adolescence with 5-year survival rates of 65-70% for localized disease and 〈 30% for de novo metastatic disease or recurrent disease. Pooled analysis of previous phase 2 trials by the Children’s Oncology Group has determined a 4-month event-free survival (EFS) of 12%. The Wee1 kinase helps regulate DNA damage repair at the G2-M checkpoint. In the presence of DNA damage, the Wee1 kinase is activated, arresting cells in the G2 phase and preventing entry into the M phase. Inhibition of the Wee1 kinase abrogates the G2-M checkpoint, forcing cancer cells to undergo unscheduled mitosis even in the presence of DNA damage, leading to mitotic catastrophe. However, the Wee1 kinase is often upregulated in OS, preserving the G2-M checkpoint and allowing tumor growth and metastases. Additionally, up to 90% of OS tumors have alterations in p53, a critical protein in the regulation of the G1-S checkpoint, especially in relapsed or refractory cases. With a dysfunctional G1-S checkpoint, cancer cells further rely on G2-M checkpoint to repair DNA damage and preserve genomic integrity. Prior studies have demonstrated that pharmacologic inhibition of the Wee1 kinase produced cell death in OS cell lines and patient-derived xenografts. While p53 mutational status appeared to modulate efficacy of the Wee1 kinase inhibitor, activity was observed in p53 wild type, mutant and null cell lines. Combination therapy studies have also been performed, demonstrating potential synergism with gemcitabine. As expected, by precipitating DNA damage, susceptibility to inhibition of the G2-M checkpoint is further increased. Methods: NCT04833582 is an ongoing, open label, multicenter, phase 1/2 clinical trial to evaluate the activity of ZN-c3, an oral Wee1 inhibitor, in combination with gemcitabine in subjects ≥12 years and ≥40 kg, with relapsed, refractory OS. Subjects are dosed once daily, continuously with ZN-c3 and receive gemcitabine 1000 mg/m 2 on days 1 and 8 of 21-day cycles. Up to 18 subjects are expected to enroll in the phase 1 portion based on a typical 3 + 3 escalation design; ̃60 subjects will be enrolled in the phase 2 portion, consisting of three stages: futility, promising clinical activity, and improved precision for clinical activity. The first two stages follow a Simon two-stage optimal design with 30 subjects, to differentiate an EFS rate at 18 weeks between 12% and 36% (which may be considered a more suitable endpoint for OS, compared with radiographic response). Tumor and skin punch biopsies are incorporated into the trial to identify potential biomarkers of treatment response. Subjects must be able to swallow oral tablets and have measurable disease by RECIST v1.1; prior exposure to gemcitabine is allowed. Global enrollment began August 1, 2021, and is ongoing. Clinical trial information: NCT04833582.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 11577-11577
    Abstract: 11577 Background: MLS is a subset of liposarcoma characterized by a translocation, (t(12;16), FUS-DDIT3 or less frequently t(12;22), EWSR1-DDIT3). Additional mutations have been reported in TERT promotor, PIK3CA, and PTEN and some were associated with the more aggressive round cell phenotype. Our study aimed at analyzing the clinical significance of these recurring mutations. Methods: MLS patients (pt) with available next-generation sequencing (NGS) data (any clinical grade) between Jan 2014 to Oct 2022 were included. Baseline characteristics, chemotherapy (CMT) response (partial response [PR] and stable disease [SD] ), and survival were collected for doxorubicin-ifosfamide (AI), doxorubicin-dacarbazine (ADIC), trabectedin (T), and eribulin (E). Binary logistic regression was used to evaluate mutations associated with response. Cox proportional-hazard regression was used to investigate associations of variables with progression-free survival (PFS) and overall survival (OS). The Kaplan-Meier method was used to estimate survival and log-rank tests were used to compare survival between groups. Results: Of 18 MLS pt included, median age at diagnosis was 42 years (R 30-65). Majority were male (12/18, 67%), had localized disease at diagnosis (13/18, 72%), and had lower extremities as primary location (10/18, 56%). Eight (44%) had round-cell 〉 5% and 15 (83%) had primary tumor 〉 5 cm. Somatic mutations were reported in 15 pt (83%); the most common were TERT promotor (61%), PTEN (28%), PIK3CA (22%), and TP53 (17%). None showed significant association with clinical factors (age, sex, size, stage, round cell 〉 5%, primary location). Forty-eight records (18 AI, 6 ADIC, 19 T, and 5 E; 3 pt received AI and T twice for different recurrences), were evaluated for PFS and 47 records had response data available. For AI, ADIC, T, and E, PR rate was 39% (7/18), 17% (1/6), 32% (6/19), and 40% (2/5), while clinical benefit (CBR: PR + SD≥ 3 mo) was 67% (12/18), 50% (3/6), 63% (12/19), and 80% (4/5), respectively. PR or CBR were not associated with the common mutations for any regimen. Median follow up (FU) from start of CMT was 4 mo (R 0.5-31). PFS (95%CI) was 11 (6.7- Not Reached [NR]), 1.2 (1.1-NR), 23 (6.3-NR), and 8 (1.3-NR) mo for AI, ADIC, T, and E, respectively. PIK3CAm (n = 15) was associated with improved PFS with CMT (PFS [95%CI] : PIK3CAwt 7.8 [6.3-11.2] vs. PIK3CAm 23 mo [1.3-NR] ; multivariate hazard ratio [HR] 0.23, p= 0.036, adjusted by gender and ADIC regimen). This appeared to be mainly driven by the PIK3CAm/ TERTwt (n = 8, HR 0.07, p= 0.006). For T, PIK3CAm/ TERTm (n = 3) showed a trend for lower PFS (1.3 mo) in contrast to other groups (23 mo). Conclusions: TERT promoter mutation was the most frequent mutation but was not associated with outcome in our small series. PIK3CAm suggested PFS benefit from CMT while, on the contrary, dual PIK3CAm/ TERTm showed a trend towards shorter PFS with T. The predictive and prognostic role of these mutations in MLS warrant further study.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 11547-11547
    Abstract: 11547 Background: Sarcomas are rare tumors of mesenchymal origin, with over 50 different histological sarcoma subtypes. The heterogeneous molecular and immunologic characteristics of sarcomas present many challenges for diagnostics and treatment plans. We aim to provide a more comprehensive molecular testing and immune profiling of sarcoma patients. Methods: The cohort consisted of samples from 200 patients diagnosed with sarcomas. WES and whole transcriptome analysis were performed on all samples. Tumor Portrait test identified genomic alterations of sarcomas and classified tumor microenvironment (TME) subtypes. The actual response to immune checkpoint inhibitor (ICI) treatment was determined using RECIST criteria and compared to response predictions by the Tumor Portrait test. Results: Across sarcoma subtypes, the most common mutations were in tumor suppressors including TP53, RB1, CDKN2A, and TSC1/2. We report fusions in 20.5% of cases, with commonly described diagnostic fusions accounting for 34/41 cases. In one case, the detection of the ZC3H7B-BCOR fusion suggested a change in diagnosis from uterine leiomyosarcoma to endometrial stromal sarcoma. Several previously unreported fusions were detected, including potentially targetable ( ARID1A-NUDC, MICAL3-MAPK) and prognostic ( YWHE-CIC) fusions. DNA damage response related genes were mutated in 12% of cases. Other less commonly detected targetable alterations were found in the following genes: MDM2, CDK4, SMARCB1, NF1, PIK3CA, NTRK1-3, FGFR1-4. Transcriptomic-based TME classification found that 52% of patients had an Immune-Enriched (IE) TME, with 26.5% and 25.5% grouped in the IE-Fibrotic and IE-non-Fibrotic subtype, respectively, which we previously showed is associated with good response to ICI treatment. In contrast, 48% of the patients presented with a Fibrotic (F, 30%) or Desert (D, 18%) subtype, are predicted to have a poor response to ICI treatment. Our classification of sarcomas based on TME subtypes conforms well with predicted responses of tumors to ICI therapy found in previous reports. Soft-tissue angiosarcomas, undifferentiated pleomorphic sarcoma, and myxofibrosarcomas presented TMEs of the IE subtype in 75% of cases, significantly more compared to 34% of the bone neoplasm cases, which are known for their favorable and unfavorable response to ICI therapy, respectively (Chi-squared test, p = 0.001). In retrospective chart review, we had response data on 24 patients who received ICI treatment. The disease control rate, defined as CR+PR+SD as best response, was significantly higher in the IE subtype compared to the F/D subtype (85.7% vs 20%, Chi-squared test, p 〈 0.003). Conclusions: Together, our findings identify actionable and diagnostic alterations in diverse mesenchymal tumors and suggest that ICI treatment may be beneficial in immune-enriched sarcomas.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Nature, Springer Science and Business Media LLC, Vol. 617, No. 7962 ( 2023-05-25), p. 764-768
    Abstract: Critical illness in COVID-19 is an extreme and clinically homogeneous disease phenotype that we have previously shown 1 to be highly efficient for discovery of genetic associations 2 . Despite the advanced stage of illness at presentation, we have shown that host genetics in patients who are critically ill with COVID-19 can identify immunomodulatory therapies with strong beneficial effects in this group 3 . Here we analyse 24,202 cases of COVID-19 with critical illness comprising a combination of microarray genotype and whole-genome sequencing data from cases of critical illness in the international GenOMICC (11,440 cases) study, combined with other studies recruiting hospitalized patients with a strong focus on severe and critical disease: ISARIC4C (676 cases) and the SCOURGE consortium (5,934 cases). To put these results in the context of existing work, we conduct a meta-analysis of the new GenOMICC genome-wide association study (GWAS) results with previously published data. We find 49 genome-wide significant associations, of which 16 have not been reported previously. To investigate the therapeutic implications of these findings, we infer the structural consequences of protein-coding variants, and combine our GWAS results with gene expression data using a monocyte transcriptome-wide association study (TWAS) model, as well as gene and protein expression using Mendelian randomization. We identify potentially druggable targets in multiple systems, including inflammatory signalling ( JAK1 ), monocyte–macrophage activation and endothelial permeability ( PDE4A ), immunometabolism ( SLC2A5 and AK5 ), and host factors required for viral entry and replication ( TMPRSS2 and RAB2A ).
    Type of Medium: Online Resource
    ISSN: 0028-0836 , 1476-4687
    RVK:
    RVK:
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2023
    detail.hit.zdb_id: 120714-3
    detail.hit.zdb_id: 1413423-8
    SSG: 11
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages