Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2020
    In:  Journal of Clinical Oncology Vol. 38, No. 15_suppl ( 2020-05-20), p. 6584-6584
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 38, No. 15_suppl ( 2020-05-20), p. 6584-6584
    Abstract: 6584 Background: Activating RAS mutations are recognized as important drivers in sporadic medullary thyroid cancer (sMTC), with a reported prevalence between 0-43%. However, few studies have looked at correlations between RAS-mutated sMTC and clinicopathologic features. Methods: Patients with sMTC diagnosed between 1992 – 2019 with NGS testing for RET and RAS mutations seen at a tertiary cancer center were retrospectively evaluated. The objective was to analyze demographic and clinical features among patients with RAS-mutated sMTC and to evaluate associations between these features and overall survival (OS). Analyses were performed to correlate patient demographics and pathologic staging with treatment characteristics, disease course, and OS. Results: We identified 42 patients (50% female) with RAS-mutated sMTC out of 218 pts with sMTC. Median age at diagnosis was 50 years (range 24-78 years). 26 (62%) patients had stage IV disease at time of diagnosis. 28 (67%) of patients had HRAS mutations and 14 (33%) had KRAS mutations. HRAS Q61R was the most common HRAS mutation type (n = 19, 45%). Median follow-up time was 64 months (range 23-274 months) during which 11 (26%) patients died. The median OS was 16.2 years, with 5- and 10- year OS of 88% and 73% respectively. Of the 20 (48%) patients who received systemic therapy, 79% had stage IV disease and tended to be older (median age 54). Median time from diagnosis to initiation of systemic therapy was 33 months. Factors associated with worse OS included distant metastases at diagnosis, shorter time interval between diagnosis and treatment, and Ctn/CEA doubling times 〈 6 months. HRAS Q61R mutations were associated with a better prognosis, with 100% 10-year OS compared with 10-year OS of 39% and 51% (p = 0.02) for other HRAS and KRAS mutations respectively. Conclusions: At a tertiary cancer center, patients with RAS-mutated sMTC had a 10-year OS rate of 73%, with significantly worse OS in patients with HRAS/KRAS mutations other than HRAS Q61R. In comparison, prior studies have reported 10-year OS rates between ~71-90% in sMTC and 10-year OS rates as low as 56% for more aggressive RET M918T sMTC mutations. The findings here are consistent with other studies that have suggested patients with RAS-mutated sMTC are at intermediate risk for aggressive disease, though there are limited data on OS rates in RAS or RAS-/RET- sMTC. Future research comparing outcomes between various RAS mutations and in comparison to RET+ and RAS-/RET- patients is needed, especially as systemic therapy use in RAS-mutated sMTC evolves.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    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. 39, No. 15_suppl ( 2021-05-20), p. 3117-3117
    Abstract: 3117 Background: Activation of the RET proto-oncogene has been identified in multiple cancer types, for example, gene rearrangements in non-small cell lung cancer (NSCLC) and papillary thyroid cancer (PTC) and activating mutations in medullary thyroid cancer (MTC), amongst others. The recent FDA approval of two highly selective RET inhibitors, selpercatinib and pralsetinib has changed the treatment paradigm of RET-driven cancers, but the significance of historical prognostic factors is unknown. Herein, we analyzed the outcomes of patients with RET-altered cancers enrolled in phase I trials and assess the utility of prognostic scores. Methods: A retrospective analysis was performed of patients treated with selective RET inhibitors at the MD Anderson Cancer Center (MDACC). Baseline clinical factors and survival data were assessed. Overall and progression free survival (OS and PFS) were estimated using the Kaplan-Meier method and multivariable Cox models were constructed. For all a p-value of 〈 0.05 was consider significant. Results: Among 126 patients, median age was 58 years (range, 15-82), most with ECOG 0-1 (n = 124). RET-mutant MTC was most frequent (n = 81), followed by RET fusion-positive NSCLC (n = 30) and RET fusion positive thyroid cancer (n = 9). RET fusion partners were KIF5B (n = 17), CCD6 (n = 12) and NCOA4 (n = 7). RET M918T mutation was the most frequent (n = 50, 63%). Most patients were treated in the relapsed/refractory (R/R) setting (n = 85) and received a median of 1 prior line of therapy (range, 0-11). Median follow up was 20 months (range, 1-43). The estimated median PFS and OS were 24 and 35 months, respectively. Overall objective response rate was 64% (81/126), 2 complete response, 79 partial response, 32 had stable disease (25%) and 13 had progressive disease (PD). The following were associated with shorter PFS and OS: age ≥50 years (p 〈 0.05), albumin 〈 4 g/dL (p 〈 0.01), brain metastases (p 〈 0.0001), hemoglobin 〈 12 g/dL ( 〈 0.05), LDH 〉 normal (p 〈 0.05), WBC ≥8 (p 〈 0.01), PD (p 〈 0.0001) and NSCLC (p 〈 0.01). The M918T mutation and ECOG 〉 0 were associated with shorter OS but not PFS (p 〈 0.05). 〉 3 metastatic sites and R/R disease were associated with inferior PFS (p = 0.04 and p = 0.01, respectively) but not OS. The Royal Marsden Hospital (RMH) and MDACC prognostic scores were significantly associated with PFS and OS (p 〈 0.01). In multivariable models including all variables significantly associated with PFS and OS (excluding LDH as this was only tested in 58 patients) albumin 〈 4 (HR 6.10, p = 0.013), brain metastases (HR 4.90, p = 0.027) and WBC ≥8 (HR 4.67, p = 0.031) were associated with inferior OS. NSCLC was significantly associated with inferior PFS (HR 5.45, p = 0.02). Conclusions: The RMH and MDACC prognostic scores predict OS in RET-aberrant cancers treated on early phase trials. Low albumin, WBC 〉 8 and brain metastases are significantly associated with inferior survival.
    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 ...
  • 3
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 33, No. 15_suppl ( 2015-05-20), p. e17012-e17012
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2015
    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. 31, No. 15_suppl ( 2013-05-20), p. TPS6100-TPS6100
    Abstract: TPS6100 Background: BRAF mutations are found in 40% of pts with newly diagnosed PTC but much more prevalent in recurrent PTC, ranging from 78-86%, suggesting they play an active role in tumor progression. Activating mutations in BRAF result in constitutive activation of MEK and subsequently ERK. ERK mediates activation of nuclear transcription factors coordinating expression of genes involved in proliferation and survival of malignant cells. PTC usually has an excellent prognosis, especially in younger pts and in pts who respond to the only effective treatment: surgery followed by radioactive iodine (RAI). However, more advanced stage disease at diagnosis, older age, and lack of RAI avidity are associated with a high rate of recurrence and distant metastasis, imparting a worse overall survival. Long-term outcomes depend highly on initial surgery outcome. Pts at highest risk of recurrence and death are those with gross residual disease after surgery and macroscopic tumor invasion. VEM is an inhibitor of the activated form of the BRAF serine-threonine kinase enzyme. The drug is FDA approved for the treatment of advanced melanoma and is currently being studied in a phase 2 trial in metastatic BRAF-mutated PTC. This is a neoadjuvant trial to determine if pharmacodynamic changes in the tumor correlate with response to drug. Methods: Pts with primary or recurrent BRAF mutated PTC who are planned for surgical resection are eligible. Pts will undergo baseline core biopsy prior to starting VEM 960mg bid. After 56 days of treatment they will undergo surgery and post-treatment specimen will be collected. Pts with widely metastatic PTC may continue VEM. The primary endpoint is to determine whether changes in ERK phosphorylation responses after treatment with VEM correlate with clinical response at day 56 in pts with locally advanced, BRAF mutated PTC. Secondary endpoints include assessments of safety of neoadjuvant VEM, response by RECIST, rate of surgical complications, persistent disease at the surgical site at 1 year, and mechanisms of resistance to VEM. The trial began enrolling pts in December 2012, with a total of 22 pts planned. This trial has no sponsor. Clinical trial information: NCT01709292.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2013
    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. 34, No. 15_suppl ( 2016-05-20), p. e17550-e17550
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2016
    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. 41, No. 16_suppl ( 2023-06-01), p. 3086-3086
    Abstract: 3086 Background: Selective RET inhibitors (RETi) have changed the paradigm for treatment of RET altered cancers. Current evidence on their efficacy comes from single-arm basket trials. In this study, we performed intrapatient efficacy comparison between approved selective RETi (selpercatinib and pralsetinib) and prior and/or subsequent therapies. We used growth modulation index (GMI) as a tool to compare efficacy, where a value of 〉 1.33 is presumed a cutoff to establish superior efficacy. Methods: We included patients with RET-positive tumors who were treated at our institution as part of RETi clinical trials. We excluded patients with no other systemic therapy received before or after the RETi of interest. We also excluded patients who received both drugs to avoid overlapping efficacy bias, and patients who were treated for ≤ 30 days or had non-evaluable disease. Since some patients have been treated beyond progression or discontinued RETi due to intolerance, we used ratios for both time to treatment takeoff (TTT) and time to progression (TTP) to compare efficacy of RETi (at the RETi treatment line (n)) to prior therapy (n-1) and to subsequent therapy (n+1). GMI was defined as the ratio between TTT/TTT (n±1) or TTP to TTP (n±1). Results: We included 66 patients who received RETi and met our inclusion criteria [39 received selpercatinib and 27 received pralsetinib]. Most patients had GMI 〉 1.33 using either TTT or TTP (61% (n=40) and 58% (n=38), respectively). The median GMI based on pre-RETi therapy was 2.1 and 1.6 (using TTT and TTP, respectively); while the median GMI based on post-RETi therapy was 4.9 and 4.4 (using TTT and TTP, respectively). Patients with GMI 〉 1.33 were more likely to have PR as best response compared to patients with GMI 〈 1.33 (85% vs 53%, p=0.005 using TTT; and 87% vs 54%, p=0.003 using TTP). GMI using TTT (n/n-1) was higher in patients with RET fusions compared to patients with RET mutations (3.7 vs 1.4, p=0.048). GMI using TTT and GMI using TTP(n/n+1) were lower in patients with WBCs 〈 8 at baseline (2.6 vs 13.8, p=0.014; 2.6 vs 11.1, p=0.014; for TTT and TTP) and GI cancer diagnosis (0.4 vs 9.3, p=0.042; 0.4 vs 10.4, p=0.042; for TTT and TTP). Conclusions: Intrapatient efficacy comparisons are feasible using GMI calculations and provide a proof of concept on the favorability of selective RETi compared to other systemic therapies. [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: 2023
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Society of Clinical Oncology (ASCO) ; 2016
    In:  Journal of Clinical Oncology Vol. 34, No. 15_suppl ( 2016-05-20), p. e17547-e17547
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 34, No. 15_suppl ( 2016-05-20), p. e17547-e17547
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2016
    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. 31, No. 15_suppl ( 2013-05-20), p. e17016-e17016
    Abstract: e17016 Background: SOR has demonstrated efficacy in phase II trials but responses are not durable. 2nd line therapy is used in clinical practice however there are no data showing clinical benefit with this approach. We sought to determine if 2nd line targeted therapy was beneficial after SOR failure. Methods: Adult pts with DTC treated with 1st line SOR for 〉 3 months were included. We compared overall survival (OS) in pts who received only 1st line SOR (grp 1) with pts treated with 2nd line therapy after 1st line SOR failure (grp 2). OS was defined as time after SOR failure until death. SOR failure was defined as discontinuation due to progression or intolerable toxicity. Results: We identified 62 DTC pts from 2006-2012 who were treated with 1st line SOR. 10 pts in grp 1 were excluded due to missing data, secondary unrelated cancers and 〈 3 mo on SOR. We included 52 pts: 31 in grp 1 and 21 in grp 2. Baseline characteristics were similar between groups (Table). FTC was more frequent in grp 2. 2nd therapy included sunitinib, pazopanib, vemurafenib, and other investigational drugs. Median time on treatment with 2nd line therapy was 14.5 mo. Median OS in grp 1 and grp 2 was 6 and 50 mo, respectively (p=0.002). After excluding pts for death 〈 3 mo after SOR end, median OS was 20 and 50 mo; there was a trend toward significance (p=0.07). Response and updated analysis will be reported at the meeting. Conclusions: Our study shows that pts receiving 2nd line therapy after SOR failure had a long OS despite more pts with FTC in this group. Acknowledging the limitations of a retrospective study, we conclude that in DTC pts, salvage targeted therapy after SOR failure appears to be beneficial and a promising strategy. [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: 2013
    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. 38, No. 15_suppl ( 2020-05-20), p. 109-109
    Abstract: 109 Background: RET gene fusions are targetable oncogenic drivers in multiple tumor types, including up to 20% of papillary thyroid cancers (PTC). Pralsetinib is an investigational, highly potent, selective inhibitor of oncogenic RET alterations. In the registration-enabling Phase 1/2 ARROW study (NCT03037385), pralsetinib demonstrated an overall response rate (ORR; response-evaluable patients [REP], central review) of 73% (19/26) in treatment-naïve patients and 61% (49/80; 2 pending confirmation) in platinum-exposed patients with RET fusion+ non-small cell lung cancer (NSCLC) and was well tolerated (data cut-off November 18, 2019). We provide an update on the clinical activity of pralsetinib in other RET fusion+ solid tumor types. Methods: ARROW consists of a phase 1 dose escalation (30–600 mg once [QD] or twice daily) followed by a phase 2 expansion (400 mg QD) in patients with advanced RET-altered solid tumors. Primary objectives were ORR and safety. Results: As of November 18, 2019, 29 patients with metastatic solid tumor types other than NSCLC (16 PTC, 1 undifferentiated thyroid, 3 pancreatic, 3 colon, 6 other) bearing a RET fusion have received pralsetinib. Efficacy data are presented for REP enrolled by July 11, 2019. In patients with thyroid cancer that is RET fusion+, ORR (investigator assessment) was 75% (9/12; all confirmed). Median (range) duration of response (DOR) was 14.5 (3.7+, 16.8) months (mo), with 67% of responding patients continuing treatment. Two patients with stable disease were continuing treatment at 11.5+ and 19.3+ mo. In other RET fusion+ cancers, ORR was 60% (3/5; all confirmed) with partial responses in 2/2 patients with pancreatic cancer (DOR 5.5, 7.4+ mo) and 1 patient with intrahepatic bile duct carcinoma (DOR 7.5 mo). Two patients with colon cancer had stable disease for 7.3 and 9.3 mo. Responses were observed across multiple fusion genotypes. In the entire safety population (all patients treated with 400 mg QD pralsetinib, regardless of diagnosis; n = 354), most treatment-related adverse events (TRAEs) were grade 1-2, and included increased aspartate aminotransferase (31%), anemia (22%), increased alanine aminotransferase (21%), constipation (21%) and hypertension (20%). Only 4% of patients in the safety population discontinued due to TRAEs. Conclusions: Pralsetinib demonstrated broad and durable antitumor activity across multiple advanced solid tumor types, regardless of RET fusion genotype, and was well tolerated. The study is ongoing and still enrolling patients in this cohort. Clinical trial information: NCT03037385.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2020
    detail.hit.zdb_id: 2005181-5
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 37, No. 15_suppl ( 2019-05-20), p. 6087-6087
    Abstract: 6087 Background: Systemic therapies targeting specific genomic alterations in advanced MTC are available or under investigation. The Afirma Genomic Sequencing Classifier (GSC) uses RNA sequencing to assess FNA specimens from cytologically indeterminate thyroid nodules, which are also tested for specific molecular aberrations associated with thyroid cancer via a suite of highly accurate malignancy classifiers. This suite can be applied independently to Bethesda V/VI nodules. The Afirma Xpression Atlas (XA) is an additional test that can be combined with Afirma GSC to report nucleotide variants and fusions across 511 cancer-associated genes. Here we report the prevalence and genomic landscape of MTC classifier positive (MTC+) FNA samples. Methods: All Afirma GSC and malignancy classifier tests run in the Veracyte Clinical Laboratory between July 2017 and January 2019 were deidentified and examined for MTC+ cases. Afirma XA was run on all such cases, and all variants and fusions were tabulated. Results: Examination of 29,895 FNAs revealed 90 MTC cases. Of 22,793 Bethesda III cases, 32 (0.14%) were MTC+. Of 5,491 Bethesda IV cases, 33 (0.60%) were MTC+. Provider-ordered testing was done on an additional 16 and 9 MTC cases from Bethesda V and VI nodules, respectively. 58% of all MTC+ samples harbored a RET variant (+/- others), 9% contained a KRAS variant (+/- others), 6% included an HRAS variant, 1% had a BRAF fusion, 1% demonstrated other fusions, and 26% held no variant/fusion. Conclusions: In our cohort, Afirma XA identified a variant or fusion in 74% of MTC+ FNAs. Currently approved or investigational therapies exist for cancers with RET, BRAF and HRAS alterations, suggesting that 64% of our series might be eligible for treatment based on genomic information from FNA. In advanced MTC, noninvasive FNA sample collection at the time of diagnosis may ultimately impact on targeted therapy selection, with the option to repeat FNA testing should the disease progress. Future studies may investigate how finding a genomic alteration by FNA can inform the management of MTC and, in the case of progressive disease, improve our understanding of the mechanisms of disease progression and drug resistance.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2019
    detail.hit.zdb_id: 2005181-5
    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