scholarly journals The value of MR-based radiomics in identifying residual disease in patients with carcinoma in situ after cervical conization

2020 ◽  
Author(s):  
Mengfan Song ◽  
Jing Lin ◽  
Fuzhen Song ◽  
Zhaoxia Qian ◽  
Guangyu Wu ◽  
...  

Abstract PurposeTo develop a magnetic resonance (MR)-based radiomics model for identifying residual disease in patients with carcinoma-in-situ (CIS) after cervical conization.Method110 patients who had CIS after conization and finally underwent hysterectomy were collected to comprise a database to establish an imaging model for predicting the residual status after conization. The imaging features were extracted from the cervical areas around the conization margin, and the performance of the imaging model was compared using different feature selection methods and with that of the pathological positive margins using receiver-operating-characteristic (ROC) analysis. Then, patients who opted for uterine preservation were included and were classified as high-risk or low-risk patients according to the radiomics model. The disease-free survival was compared between the different risk groups using the Kaplan-Meier curve.ResultsThe model with Boruta features achieved an area under the curve (AUC) of 0.889 and an accuracy of 87.3% in the test cohort and significantly outperformed the model created with the random forest method, which had an AUC of 0.701 (p = 0.039), and the positive margins (p = 0.004). Further validation with patients with uterine preservation showed that the patients classified as high risk with the radiomics model were more likely to have tumor recurrence/residual disease in the follow-up period than low-risk patients(p = 0.024).ConclusionsRadiomics can be used to identify residual disease in patients with CIS after cervical conization and could have the potential to predict recurrence in patients who opt for uterine preservation.

2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Mengfan Song ◽  
Jing Lin ◽  
Fuzhen Song ◽  
Dan Wu ◽  
Zhaoxia Qian

AbstractCarcinoma in situ (CIS) of the uterine cervix is a precursor to cervical carcinoma. However, hysterectomy can be avoided in patients who can be treated by cone biopsy. Previous studies have shown that imaging-based approaches allow for the noninvasive visualization of cervical cancer, and radiomics has high accuracy in classifying cancer and predicting treatment outcome for different cancer types. To develop a magnetic resonance (MR)-based radiomics model for identifying residual disease in patients with CIS after cervical conization. Patients who had CIS after conization and finally underwent hysterectomy were collected to comprise a database to establish an imaging model for predicting the residual status after conization. Then, patients who opted for uterine preservation were classified as high-risk or low-risk patients according to the model. The disease-free survival was compared between the different risk groups using the Kaplan–Meier curve. The model built with the Boruta features outperformed the random forest model. Further validation with patients with uterine preservation showed that the patients classified as high risk were more likely to have tumor recurrence/residual disease in the follow-up period. In conclusion, radiomics can be used to identify residual disease in patients with CIS after cervical conization and could have the potential to predict recurrence in patients who opt for uterine preservation.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e12046-e12046 ◽  
Author(s):  
Yao Yuan ◽  
Alison Len Van Dyke ◽  
Allison W. Kurian ◽  
Serban Negoita ◽  
Valentina I. Petkov

e12046 Background: OncotypeDX DCIS is a 12-gene assay designed to predict the 10-year risk of local recurrence and to guide treatment decisions, specifically the benefit of radiation therapy in breast ductal carcinoma in situ (DCIS). The test became available in December 2011 and is not currently recommended by guidelines. The Surveillance, Epidemiology and End Results (SEER) program captures cancer data at the population-level and has been conducting annual linkages with Genomic Health Clinical Laboratory, the only lab performing the test, to identify patients receiving the test. Methods: SEER cases diagnosed with in situ breast cancer (DCIS or papillary in situ) between 2011-2015 were included in the analysis. SEER data on patient demographics, tumor characteristics, and treatments were combined with linkage variables for OncotypeDX DCIS tests reported by Genomic Health. Logistic regression was used to identify which patient related factors were associated with having received the test and to evaluate the relationship between test generated risk categories and treatments. Results: Of the 68,826 in situ breast cancer cases, 2,155 were linked to DCIS test data. Test utilization increased from < 1% to 5.3% for patients diagnosed in 2011 vs. 2015. Patients were less likely to receive the test if they had larger and higher-grade tumors, were divorced, had Medicaid insurance, and were in the lowest socioeconomic status tertile. The majority of patients (68%) were at low risk, 17% intermediate, and 15% in the high risk group. Patients at intermediate or high risk were more likely to receive radiation (OR = 2.4, 95% CI: 1.8-3.2 and OR = 3, 95% CI: 2.3,4.1, respectively) than the low risk group. High risk patients were more likely than low risk patients to receive chemotherapy (OR = 4.3, 95% CI: 1.2, 14.4) and to undergo mastectomy than lumpectomy (OR = 1.47, 95% CI: 1.12-1.93). Conclusions: Clinical adoption of the OncotypeDX DCIS test has been slow. The association between multiple demographic factors and receiving the test indicated disparities in the US population. Clinical factors also influenced whether patients received the test. OncotypeDX DCIS results appeared to guide clinical decisions.


PLoS ONE ◽  
2019 ◽  
Vol 14 (6) ◽  
pp. e0217562 ◽  
Author(s):  
João Paolo Bilibio ◽  
Heleusa Ione Monego ◽  
Márcia Luiza Appel Binda ◽  
Ricardo dos Reis

1992 ◽  
Vol 10 (4) ◽  
pp. 599-605 ◽  
Author(s):  
D C Allred ◽  
G M Clark ◽  
A K Tandon ◽  
R Molina ◽  
D C Tormey ◽  
...  

PURPOSE Amplification and/or overexpression of the HER-2/neu oncogene have been shown to correlate with poor clinical outcome in patients with axillary node-positive breast cancer. In contrast, the prognostic significance of HER-2/neu in node-negative disease is controversial. This study was undertaken to evaluate further the relationship between HER-2/neu and clinical outcome in node-negative disease. PATIENTS AND METHODS Overexpression of HER-2/neu was evaluated by permanent-section immunohistochemistry in tumors from 613 patients with long-term clinical follow-up enrolled in the Intergroup Study 0011. Patients were stratified into low-risk (n = 307) and high-risk (n = 306) groups on the basis of tumor size and estrogen-receptor (ER) status. Low-risk patients were defined as having small (less than 3 cm), ER-positive tumors and were observed without additional treatment after initial surgery. High-risk patients had either ER-negative or large (greater than or equal to 3 cm), ER-positive tumors and were randomized to be observed (n = 146) or to receive adjuvant chemotherapy (n = 160) after surgery. RESULTS The rate of HER-2/neu overexpression was 14.3% in all tumors combined and was higher in invasive carcinomas with (21.5%) than without (11.2%) a significant noninvasive or in situ histologic component (P less than .0001). There was no relationship between overexpression and clinical outcome in the natural history setting of combined low-risk and high-risk patients not receiving adjuvant therapy (n = 453). Based on the reasoning that the influence of HER-2/neu may have been obscured by high-risk features and/or the presence of noninvasive carcinoma, we also analyzed the subset of patients with low-risk lesions not containing a significant in situ component (n = 179). Patients of this group with HER-2/neu-positive tumors showed only 40% disease-free survival (DFS) at 5 years, compared with over 80% in patients with HER-2/neu-negative tumors (P less than .0001). A similar inverse correlation was observed between overexpression and overall survival in the same group of patients (P = .0001). In a separate analysis involving patients receiving adjuvant chemotherapy, those with HER-2/neu-negative tumors showed significantly improved DFS in response to therapy compared with patients with HER-2/neu-positive tumors. CONCLUSION Overexpression of HER-2/neu is associated with poor clinical outcome in a subset of node-negative patients with small, ER-positive, predominantly invasive tumors and may play a role in resistance to adjuvant chemotherapy.


Diagnostics ◽  
2021 ◽  
Vol 11 (10) ◽  
pp. 1889
Author(s):  
Linda J. Hong ◽  
Sandy Huynh ◽  
Joy Kim ◽  
Laura Denham ◽  
Mazdak Momeni ◽  
...  

Background: Adenocarcinoma in situ (AIS) of the cervix, is increasing in incidence, particularly in women of reproductive age. Fertility preservation is often desired. In a predominantly Hispanic population, we sought to determine the incidence of occult cervical cancer co-existing with AIS, and evaluate how conization margin status correlates with residual disease upon hysterectomy. Methods: A retrospective study utilizing a comprehensive cancer center database was conducted. Data from patients with histologically proven AIS of the cervix were abstracted. Results: Of 47 patients that met the criteria, 23 (49%) were Hispanic, 21 (45%) were White, two (4%) were Asian, and one (2%) was Black. The median age was 37. Forty-two patients underwent cervical conizations; 13/42 (48%) had positive margins upon conization; 28/42 (67%) underwent hysterectomies. Furthermore, 6/13 (46%) patients with positive conization margins had residual disease in hysterectomy specimens, with 2/13 (15%) found to have invasive cancer. In contrast, 0/14 (0%) of patients with negative margins had residual disease (p = 0.036, Chi-squared 4.41, df = 1). In total, 2/27 (7%) patients who underwent hysterectomies had invasive cancer (7%). Conclusions: Positive margins upon cervical conization for AIS of the cervix were correlated with a relatively high rate of residual AIS and occult invasive cancer. Negative conization margins were correlated with no residual disease. Those patients may be candidates for fertility-sparing treatment.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2739-2739 ◽  
Author(s):  
Richard Dillon ◽  
Robert K. Hills ◽  
Sylvie D Freeman ◽  
Nicola Potter ◽  
Adam Ivey ◽  
...  

Abstract Introduction Relapse remains the most common cause of treatment failure for patients with acute myeloid leukaemia (AML) who undergo allogeneic stem cell transplantation (SCT) and carries a grave prognosis. Multiple studies have identified the presence of minimal residual disease (MRD) prior to SCT assessed by flow cytometry (FCM) as a strong predictor of relapse: however, little is known about the impact of molecular MRD pre-SCT. Molecular techniques can provide 100-1000 fold greater sensitivity than FCM in NPM1 mutated AML allowing identification of patients with very low level MRD prior to SCT. Peri-transplant management decisions for patients with positive molecular MRD are highly challenging due to the absence of robust outcome data. Methods Between 2009-14 the NCRI AML17 study enrolled 3215 adult non-APML patients aged 16-77 eligible for intensive chemotherapy. Central screening for NPM1 mutations was positive in 861/2949 (29%); 530 patients provided serial samples for MRD monitoring. Paired blood (PB) and bone marrow aspirates (BM) were requested after each chemotherapy cycle and then quarterly; additional samples were requested pre- and post-SCT. Samples were analysed by RT-qPCR using a suite of 27 mutation-specific reverse primers. Results were only fed back to clinicians after June 2012 when patients could be treated for confirmed re-emergent or persistent molecular positivity. Post-remission treatment was determined according to the validated NCRI risk score, with poor-risk patients recommended for SCT during first complete remission (CR1). Survival analysis was performed using the logrank test. Results In total 108/530 patients received SCT (CR1 57 (52%), after molecular relapse or progression (MR) 30 (28%), CR2 21 (19%)). Five-year survival post-SCT (5y-OS) was 65% in CR1, and 54% in MR/CR2 (p=0.3). After MR 26/30 patients received chemotherapy prior to SCT. Evaluable pre-SCT PB and BM samples taken within 60 days of SCT were available for 104 and 78 patients (both available n=74). Considering PB samples, 5y-OS was 73% (median OS (mOS) not reached (NR)) for MRD-ve patients (n=74) vs. 30% (mOS 8.1 m) for any PB positivity (n=30) (p<0.0001). Patients with a negative pre-SCT BM had 5y-OS of 79% (mOS NR) vs 47% (mOS 11.9 m) if the BM was positive (p=0.002). Of the 47 patients who received additional chemotherapy for morphologic or molecular relapse or progression, 26 (55%) converted to MRD negativity accounting for 44% of 59 patients who were MRD negative pre-SCT. Ninety-three per cent of 59 pre SCT-MRD negative patients had been MRD negative in the PB after the second cycle of induction (a previously identified marker of favourable outcome). A threshold of 200 mutant NPM1 transcripts/105 ABL copies in the pre-SCT PB sample split patients into 3 groups with 2y-OS of 81% (negative, n=74), 54% (low, n=13) and 9% (high, n=17; p<0.0001). In the BM a threshold of 1000 copies defined 3 groups with a 2y-OS of 86% (negative, n=36), 56% (low, n=32) and 12.5% (high, n=8; p<0.0001). Thirty four patients were positive for FLT3-ITD at diagnosis (5y-OS 55%) and 74 were negative (5y-OS 62%; p=0.3). For patients without FLT3 ITD, negative, low and high levels of MRD in the pre-SCT PB sample were associated with 2y-OS of 79% (n=53), 88% (n=8) and 0% (n=9) (p<0.0001). For BM samples, 2y-OS was 78% (n=28), 62% (n=24) and 0% (n=5; p=0.0001). For patients with FLT3 ITD, corresponding 2y-OS for PB samples were 89% (n=21), 0% (n=5) and 25% (n=8; p=0.0005); for BM samples 2y-OS was 83% (n=9), 38% (n=8) and 25% (n=4; p=0.02). We assigned 80 patients to high and low risk groups based on FLT3 ITD status and pre-SCT MRD level in PB and BM. High-risk patients (FLT3 ITD and any detectable level of MRD in the PB or BM, or FLT3 ITD negative with > 200 copies in the PB or > 1000 copies in the BM) had 3y-OS of 14% (n=27, mOS 6.5 months) compared with 80% for low-risk patients (n=53, p<0.0001) (fig 1). Conclusions Patients who test negative for NPM1 mutant transcripts immediately before SCT have a favourable outcome which is also observed in FLT3 ITD negative patients who are MRD positive at levels which do not exceed 200 copies in the PB or 1000 copies in the BM. FLT3 ITD mutated patients with any level of MRD prior to SCT, and FLT3 ITD negative patients with transcript levels above these thresholds, have a very high risk of relapse and may benefit from further chemotherapy or FLT3 inhibition prior to or after SCT; studies to investigate this are urgently needed. Disclosures Hills: Daiichi Sankyo: Consultancy, Honoraria. Cavenagh:Celgene: Honoraria, Research Funding, Speakers Bureau; Amgen: Honoraria, Speakers Bureau; Janssen: Honoraria, Speakers Bureau; Takeda: Research Funding, Speakers Bureau; Novartis: Honoraria, Speakers Bureau. Russell:Daiichi Sankyo: Consultancy; Pfizer: Consultancy, Honoraria, Speakers Bureau; Jazz Pharma: Speakers Bureau.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 496-496 ◽  
Author(s):  
Ajay J. Vora ◽  
Chris Mitchell ◽  
Nicholas Goulden ◽  
Sue Richards

Abstract Abstract 496 UKALL 2003 is an ongoing randomised controlled trial investigating treatment modification according to Minimal Residual Disease (MRD) status at the end of induction and week 11 in children and young adults (up to age 25 years) with ALL. Whilst the randomisation testing the benefit of intensification for an MRD defined high risk group remains open, a treatment reduction randomisation for MRD low risk patients closed in August 2009 after recruiting the target number of patients. Over 2600 patients have so far been enrolled into the trial and we have observed a significant improvement in 5 year event-free (EFS) compared with its predecessor, ALL 97/99, (EFS 87 vs 80%, p<0.0005), most probably related to use of dexamethasone and pegylated asparaginase in all patients. Here we report the results of the MRD low risk randomisation. Patients in the trial are initially stratified by NCI risk, blast karyotype, and morphological marrow response at day 8/15 of induction for allocation to one of 3 treatment regimens, escalating in intensity (A, B, C). NCI standard risk (SR) patients receive a 3 drug induction (Regimen A) whilst high risk (HR) patients receive 4 drugs including daunorubicin (Regimen B). Patients with a slow early response at day 8 (NCI HR) or day 15 (NCI SR) of induction and patients with poor risk cytogenetics are transferred to Regimen C. Patients without poor risk cytogenetics and who have a rapid early response are eligible for the MRD randomisations. MRD was measured using Real Time Quantitative PCR with a sensitivity of 10−4. Five hundred and twenty-one patients who were either MRD negative at day 29 of induction or positive < 10−4 at day 29 but negative by week 11 entered a randomisation comparing two courses of delayed intensification (standard arm, n = 261) with one (treatment reduction arm, n = 260). Of these patients, 342 (65%) were NCI SR and 179 (35%) NCI HR. With follow-up to October 2009 (median 2 years 9 months), there is no difference in 5 year EFS for patients in the standard (94%) or reduced treatment arms (96%). As shown in table 1, the number of patients with relapse is similar in both arms but there was a non-significant excess of deaths in remission, serious adverse events and grade 3/4 toxicity in recipients of 2 DIs. 7/10 relapses have involved the CNS. The 3 CR deaths in the 2 DI arm were due to gram negative sepsis (DI1), pulmonary haemorrhage (DI2) and RSV pneumonitis (Maintenance, Down). There have been no relapses or CR deaths among 23 MRD low risk patients in the 16 – 25 age group. Table 1 1DI 2DI Statistics for 2DI Obs/N Obs/N Odds ratio(95% CI) 2p Any event 6/260 7/261 1.19 (0.40–3.53) 0.76 Relapse 6/260 4/261 0.69 (0.20–2.38) 0.57 Remission death 0/260 3/261 7.40 (0.77–71.14) 0.08 Grade 3/4 toxicity 177 (68%) 191 (71%) 0.2 SAEs 64 (25%) 77 (29.5%) 0.2 This randomised study provides evidence that treatment can be reduced for around 40% of children and young adults who show rapid clearance of MRD by the end of induction. These patients have an excellent outcome with < 5% risk of relapse on a treatment protocol that does not include cranial radiotherapy or high dose methotrexate and gives minimal exposure to anthracyclines and alkylating agents. Future trials should test the feasibility of further reductions in treatment for this group of patients. Disclosures: No relevant conflicts of interest to declare.


2014 ◽  
Vol 132 (1) ◽  
pp. 76-80 ◽  
Author(s):  
Elena S. Diaz ◽  
Chisa Aoyama ◽  
Mary Anne Baquing ◽  
Anna Beavis ◽  
Elvio Silva ◽  
...  

2020 ◽  
Vol 13 (1) ◽  
Author(s):  
Katja Weisel ◽  
Andrew Spencer ◽  
Suzanne Lentzsch ◽  
Hervé Avet-Loiseau ◽  
Tomer M. Mark ◽  
...  

Abstract Background Multiple myeloma (MM) patients with high cytogenetic risk have poor outcomes. In CASTOR, daratumumab plus bortezomib/dexamethasone (D-Vd) prolonged progression-free survival (PFS) versus bortezomib/dexamethasone (Vd) alone and exhibited tolerability in patients with relapsed or refractory MM (RRMM). Methods This subgroup analysis evaluated D-Vd versus Vd in CASTOR based on cytogenetic risk, determined using fluorescence in situ hybridization and/or karyotype testing performed locally. High-risk patients had t(4;14), t(14;16), and/or del17p abnormalities. Minimal residual disease (MRD; 10−5 sensitivity threshold) was assessed via the clonoSEQ® assay V2.0. Of the 498 patients randomized, 40 (16%) in the D-Vd group and 35 (14%) in the Vd group were categorized as high risk. Results After a median follow-up of 40.0 months, D-Vd prolonged median PFS versus Vd in patients with standard (16.6 vs 6.6 months; HR, 0.26; 95% CI, 0.19-0.37; P < 0.0001) and high (12.6 vs 6.2 months; HR, 0.41; 95% CI, 0.21–0.83; P = 0.0106) cytogenetic risk. D-Vd achieved deep responses, including higher rates of MRD negativity and sustained MRD negativity versus Vd, regardless of cytogenetic risk. The safety profile was consistent with the overall population of CASTOR. Conclusion These updated data reinforce the effectiveness and tolerability of daratumumab-based regimens for RRMM, regardless of cytogenetic risk status. Trial registration ClinicalTrials.gov, NCT02136134. Registered 12 May 2014


Sign in / Sign up

Export Citation Format

Share Document