Camelia Sima
Memorial Sloan Kettering Cancer Center
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Camelia Sima.
Journal of Thoracic Oncology | 2013
Farhood Farjah; Feiran Lou; Camelia Sima; Valerie W. Rusch; Nabil P. Rizk
Introduction: Guidance is limited for invasive staging in patients with lung cancer without mediastinal disease by positron emission tomography (PET). We developed and validated a prediction model for pathologic N2 disease (pN2), using six previously described risk factors: tumor location and size by computed tomography (CT), nodal disease by CT, maximum standardized uptake value of the primary tumor, N1 by PET, and histology. Methods: A cohort study (2004–2009) was performed in patients with T1/T2 by CT and N0/N1 by PET. Logistic regression analysis was used to develop a prediction model for pN2 among a random development set (n = 625). The model was validated in both the development set, which comprised two thirds of the patients and the validation set (n = 313), which comprised the remaining one third. Model performance was assessed in terms of discrimination and calibration. Results: Among 938 patients, 9.9% had pN2 (9 detected by invasive staging and 84 intraoperatively). In the development set, univariate analyses demonstrated a significant association between pN2 and increasing tumor size (p < 0.001), nodal status by CT (p = 0.007), maximum standardized uptake value of the primary tumor (p = 0.027), and N1 by PET (p < 0.001); however, only N1 by PET was associated with pN2 (p < 0.001) in the multivariate prediction model. The model performed reasonably well in the development (c-statistic, 0.70; 95% confidence interval, 0.63–0.77; goodness of fit p = 0.61) and validation (c-statistic, 0.65; 95% confidence interval, 0.56–0.74; goodness-of-fit p = 0.19) sets. Conclusion: A prediction model for pN2 based on six previously described risk factors has reasonable performance characteristics. Observations from this study may guide prospective, multicenter development and validation of a prediction model for pN2.
Annals of Oncology | 2016
Bob T. Li; Alexander Drilon; Melissa Lynne Johnson; Meier Hsu; Camelia Sima; C. McGinn; Hirofumi Sugita; Mark G. Kris; Christopher G. Azzoli
BACKGROUNDnWhile previous studies have reported on the prognostic value of total plasma cell-free deoxyribonucleic acid (cfDNA) in lung cancers, few have prospectively evaluated its predictive value for systemic therapy response.nnnPATIENTS AND METHODSnWe conducted a prospective study to evaluate the association between changes in total cfDNA and radiologic response to systemic therapy in patients with stage IIIB/IV non-small-cell lung cancers (NSCLCs). Paired blood collections for cfDNA and computed tomography (CT) assessments by RECIST v1.0 were performed at baseline and 6-12 weeks after therapy initiation. Total cfDNA levels were measured in plasma using quantitative real-time polymerase chain reaction. Associations between changes in cfDNA and radiologic response, progression-free survival (PFS), and overall survival (OS) were measured using Kruskal-Wallis and Kaplan-Meier estimates.nnnRESULTSnA total of 103 patients completed paired cfDNA and CT response assessments. Systemic therapy administered included cytotoxic chemotherapy in 57% (59/103), molecularly targeted therapy in 17% (17/103), and combination therapy in 26% (27/103). Median change in cfDNA from baseline to response assessment did not significantly differ by radiologic response categories of progression of disease, stable disease and partial response (P = 0.10). However, using radiologic response as continuous variable, there was a weak positive correlation between change in radiologic response and change in cfDNA (Spearmans correlation coefficient 0.21, P = 0.03). Baseline cfDNA levels were not associated with PFS [hazard ratio (HR) = 1.06, 95% confidence interval (CI) 0.93-1.20, P = 0.41] or OS (HR = 1.04, 95% CI 0.93-1.17, P = 0.51), neither were changes in cfDNA.nnnCONCLUSIONSnIn this large prospective study, changes in total cfDNA over time did not significantly predict radiologic response from systemic therapy in patients with advanced NSCLC. Pretreatment levels of total cfDNA were not prognostic of survival. Total cfDNA level is not a highly specific predictive biomarker and future investigations in cfDNA should focus on tumor-specific genomic alterations using expanded capabilities of next-generation sequencing.
PLOS ONE | 2011
Elliot L. Servais; Kei Suzuki; Christos Colovos; Luis Rodriguez; Camelia Sima; Martin Fleisher; Valerie W. Rusch; Michel Sadelain; Prasad S. Adusumilli
Tumor biomarkers provide a quantitative tool for following tumor progression and response to therapy. However, investigations of clinically useful tumor biomarkers are time-consuming, costly, and limited by patient and tumor heterogeneity. In addition, assessment of biomarkers as indicators of therapy response is confounded by the concomitant use of multiple therapeutic interventions. Herein we report our use of a clinically relevant orthotopic animal model of malignant pleural mesothelioma for investigating tumor biomarkers. Utilizing multi-modality imaging with correlative histopathology, we demonstrate the utility and accuracy of the mouse model in investigating tumor biomarkers – serum soluble mesothelin-related peptide (SMRP) and osteopontin (OPN). This model revealed percentage change in SMRP level to be an accurate biomarker of tumor progression and therapeutic response – a finding consistent with recent clinical studies. This in vivo platform demonstrates the advantages of a validated mouse model for the timely and cost-effective acceleration of human biomarker translational research.
Annals of Oncology | 2016
Alexander Drilon; Isabella Bergagnini; Lukas Delasos; Joshua K. Sabari; Kaitlin M. Woo; Andrew J. Plodkowski; Lu Wang; Matthew D. Hellmann; P. Joubert; Camelia Sima; Roger S. Smith; Romel Somwar; Natasha Rekhtman; M. Ladanyi; Gregory J. Riely; Mark G. Kris
BACKGROUNDnRET rearrangements are targetable, oncogenic lung cancer drivers. While previous series have shown durable clinical benefit with pemetrexed-based therapies in ALK- and ROS1-rearranged lung cancers, the benefits of pemetrexed-based treatments in patients with RET-rearranged lung cancers relative to other genomic subsets have not previously been explored.nnnPATIENTS AND METHODSnA retrospective review of patients with pathologically confirmed stage IIIB/IV lung adenocarcinomas and evidence of a RET, ROS1, or ALK rearrangement, or a KRAS mutation was conducted. Patients were eligible if they received treatment with pemetrexed alone or in combination. The primary outcome of progression-free survival (PFS), and secondary outcomes of overall response rate (ORR, RECIST v1.1), time to progression (TTP), and time to treatment discontinuation were compared between RET-rearranged and groups of ROS1-rearranged, ALK-rearranged, and KRAS-mutant lung cancers.nnnRESULTSnWe evaluated 104 patients. Patients with RET-rearranged lung cancers (n = 18) had a median PFS of 19 months [95% confidence interval (CI) 12-not reached (NR)] that was comparable with patients with ROS1- (23 months, 95% CI 14-NR, n = 10) and ALK-rearranged (19 months, 95% CI 15-36, n = 36) lung cancers, and significantly improved compared with patients with KRAS-mutant lung cancers (6 months, 95% CI 5-9, P < 0.001, n = 40). ORR (45%), median TTP (20 months, 95% CI 17-NR), and median time to treatment discontinuation (21 months, 95% CI 6-NR) in patients with RET-rearranged lung cancers were not significantly different compared with patients with ALK- and ROS1-rearranged lung cancers, and improved compared with patients with KRAS-mutant lung cancers.nnnCONCLUSIONnDurable benefits with pemetrexed-based therapies in RET-rearranged lung cancers are comparable with ALK- and ROS1-rearranged lung cancers. When selecting therapies for patients with RET-rearranged lung cancers, pemetrexed-containing regimens should be considered.
Cancer Biomarkers | 2014
Emil Lou; Melissa Lynne Johnson; Camelia Sima; Rita Gonzalez-Espinoza; Martin Fleisher; Mark G. Kris; Christopher G. Azzoli
BACKGROUNDnSerum biomarkers are not in routine clinical use for diagnosis, prognosis, or treatment selection in lung cancer.nnnOBJECTIVEnWe examined serum protein biomarkers from patients with metastatic lung cancer to determine whether they correlate with progression-free survival (PFS), overall survival (OS), or histologic subtype.nnnMETHODSnSerum samples were collected prior to chemotherapy from 153 patients with metastatic lung cancer treated at Memorial Sloan-Kettering Cancer Center. Serum biomarkers were selected for ELISA testing based on their availability in a CLIA-certified clinical laboratory: ProGRP, SCC-Ag, NSE, CYFRA 21-1, TIMP1, and HE4. Pretreatment biomarker levels were correlated with outcome using proportional hazards analysis and tumor histology using logistic regression analysis.nnnRESULTSnUnivariate analysis indicated that only higher levels of CYFRA 21-1 were significantly associated with worsened PFS (HR 1.3, 95% CI 1.1--1.5, p< 0.01) and OS (HR 1.4, 95% CI 1.2-1.7, p< 0.001). Multivariate analysis of NSE, CYFRA 21-1, and TIMP1 indicated that CYFRA 21-1 remained independently associated with lower OS (HR 1.3, 95% CI 1.1-1.6, p< 0.01). Univariate analysis indicated that ProGRP (OR 3.3, 95% CI 1.7-6.5, p< 0.001) and NSE (OR 4.8, 95% CI 2.6-8.8, p< 0.0001) had the highest probabilities of differentiating SCLC from NSCLC. Multivariate analysis of these two markers demonstrated that they predicted SCLC histology with 94% accuracy. Univariate analysis showed that only SCCL-Ag distinguished squamous cell histology from adenocarcinoma (OR 4.4, 95% CI 1.7-11.5, p< 0.01).nnnCONCLUSIONSnSerum CYFRA 21-1 may be useful in predicting patient survival, and serum ProGRP, NSE 21-1, and SCCL-Ag may be helpful in distinguishing between lung cancer sub-types.
PLOS ONE | 2016
Bob T. Li; Emil Lou; Meier Hsu; Helena A. Yu; Jarushka Naidoo; Marjorie Glass Zauderer; Camelia Sima; Melissa Lynne Johnson; Mariza Daras; Lisa M. DeAngelis; Martin Fleisher; Mark G. Kris; Christopher G. Azzoli
Background Lung cancers account for the majority of brain metastases which pose major therapeutic challenges. Biomarkers prognosticating for the development of brain metastases in patients with non-small cell lung cancers (NSCLC) may improve personalized care. Six serum proteomic biomarkers were previously investigated at Memorial Sloan Kettering but their associations with brain metastases were unknown. Methods Serum NSE, CYFRA 21–1, ProGRP, SCC-Ag, TIMP1, and HE4 by ELISA-based proteomic assays were prospectively collected from consecutive patients with stage IV NSCLC. Pre-treatment serum biomarker levels as well as age, histology, and epidermal growth factor receptor (EGFR) mutation status were evaluated for association with the baseline presence of brain metastases using logistic regression and multivariable analysis. For patients without brain metastases at baseline, the cumulative incidence of subsequent brain metastases were compared according to baseline biomarkers and clinical factors using Gray’s test. Results A total of 118 patients were enrolled, 31 (26%; 95% CI 0.19–0.35) had brain metastases at baseline and a further 26 (22%; 95% CI 0.15–0.30) developed brain metastases subsequently. Pre-treatment serum biomarker levels were available in 104 patients. There was no significant association between the six serum biomarkers and the baseline presence or subsequent development of brain metastases. Age younger than 65 years was the only clinical factor significantly associated with brain metastasis at baseline (OR 3.00; 95% CI 1.22–7.34, P = 0.02) by multivariable analysis. A trend toward increased cumulative incidence of subsequent brain metastases was observed in patients with EGFR mutation (p = 0.2), but this was not statistically significant possibly due to small sample size. Conclusions Serum NSE, CYFRA 21–1, Pro-GRP, SCC-Ag, TIMP1, and HE4 are not significantly associated with brain metastases. Our methods taking into account follow-up time may be applied to independent datasets to identify a patient cohort with a higher biologic propensity for developing brain metastases. Such information may be useful for the study of agents targeting the development of brain metastases.
Annals of Oncology | 2016
A. L. Ho; L. Dunn; Eric J. Sherman; Matthew G. Fury; Shrujal S. Baxi; R. Chandramohan; S. Dogan; Luc G. T. Morris; G. D. Cullen; S. Haque; Camelia Sima; Andy Ni; Cristina R. Antonescu; Nora Katabi; D. G. Pfister
BACKGROUNDnRecurrent/metastatic adenoid cystic carcinoma (ACC) is an incurable disease with no standard treatments. The majority of ACCs express the oncogenic transcription factor MYB (also c-myb), often in the context of a MYB gene rearrangement. This phase II trial of the tyrosine kinase inhibitor (TKI) axitinib (Pfizer) tested the hypothesis that targeting pathways activated by MYB can be therapeutically effective for ACC.nnnPATIENTS AND METHODSnThis is a minimax two-stage, phase II trial that enrolled patients with incurable ACC of any primary site. Progressive or symptomatic disease was required. Patients were treated with axitinib 5 mg oral twice daily; dose escalation was allowed. The primary end point was best overall response (BOR). An exploratory analysis correlating biomarkers to drug benefit was conducted, including next-generation sequencing (NGS) in 11 patients.nnnRESULTSnThirty-three patients were registered and evaluable for response. Fifteen patients had the axitinib dose increased. Tumor shrinkage was achieved in 22 (66.7%); 3 (9.1%) had confirmed partial responses. Twenty-five (75.8%) patients had stable disease, 10 of whom had disease stability for >6 months. The median progression-free survival (PFS) was 5.7 months (range 0.92-21.8 months). Grade 3 axitinib-related toxicities included hypertension, oral pain and fatigue. A trend toward superior PFS was noted with the MYB/NFIB rearrangement, although this was not statistically significant. NGS revealed three tumors with 4q12 amplification, producing increased copies of axitinib-targeted genes PDGFR/KDR/KIT. Two 4q12 amplified patients achieved stable disease for >6 months, including one with significant tumor reduction and the longest PFS on study (21.8 months).nnnCONCLUSIONSnAlthough the primary end point was not met, axitinib exhibited clinical activity with tumor shrinkage achieved in the majority of patients with progressive disease before trial enrollment. Analysis of MYB biomarkers and genomic profiling suggests the hypothesis that 4q12 amplified ACCs are a disease subset that benefit from TKI therapy.
The Annals of Thoracic Surgery | 2015
Usman Ahmad; Haoxian Yang; Camelia Sima; Daniel Buitrago; R. Taylor Ripley; Kei Suzuki; Manjit S. Bains; Nabil P. Rizk; Valerie W. Rusch; James Huang; Prasad S. Adusumilli; Gaetano Rocco; David R. Jones
BACKGROUNDnWe sought to determine the prognostic variables associated with overall survival (OS) and recurrence-free probability (RFP) in patients with primary and secondary sternal tumors treated with surgical resection.nnnMETHODSnA retrospective analysis of patients who underwent resection of primary or secondary sternal tumors at 2 cancer institutes between 1995 and 2013 was performed. OS and RFP were estimated using the Kaplan-Meier method, and predictors of OS and RFP were analyzed using the Cox proportional hazards model.nnnRESULTSnSternal resection was performed in 78 patients with curative (67 [86%]) or palliative (6 [8%]) intent. Seventy-three patients (94%) had malignant tumors, of which 28 (36%) were primary and 45 (57%) were secondary malignancies. Sternal resections were complete in 13 patients (17%) and partial in 65 (83%). There were no perioperative deaths, and grade III/IV complications were noted in 17 patients (22%). The 5-year OS was 80% for patients with primary malignant tumors, 73% for patients with nonbreast secondary malignant tumors, and 58% for patients with breast tumors (p = 0.85). In the overall cohort, R0 resection was associated with prolonged 5-year OS (84% vs 20%) on univariate (p = 0.004) and multivariate (adjusted hazard ratio, 3.37; p = 0.029) analysis. On subgroup analysis, R0 resection was associated with improved OS and RFP only for patients with primary malignant tumors.nnnCONCLUSIONSnSternal resection can achieve favorable OS for patients with primary and secondary sternal tumors. R0 resection is associated with improved 5-year OS and RFP in patients with primary malignant tumors. We did not detect a similar effect in patients with breast or nonbreast secondary tumors.
Cancer Research | 2015
Bob T. Li; Marjorie G. Zauderer; Jamie E. Chaft; Alexander Drilon; Juliana Eng; Camelia Sima; Vicky Makker; Gopa Iyer; Yelena Y. Janjigian; David M. Hyman; Maria E. Arcila; José Baselga; Mark G. Kris
Background: The use of therapies targeting the human epidermal growth factor receptor 2 (HER2, ERBB2) has transformed care in breast and gastric cancers. HER2 amplification has emerged as a therapeutic target in 2-5% of lung cancers, 6% of bladder cancers, 5-12% of endometrial cancers, and 2-5% of ovarian and colorectal cancers. High level HER2 protein overexpression by immunohistochemistry correlates with HER2 amplification. Ado-trastuzumab emtansine is an antibody drug conjugate linking the HER2 targeted monoclonal antibody trastuzumab, with the cytotoxic anti-microtubule drug emtansine. This agent improves response and survival in patients with HER2 amplified or HER2 overexpressed breast cancers. We hypothesize that ado-trastuzumab emtansine will be effective in any tumor with HER2 amplification or overexpression, regardless of the primary site. Methods/Design: This phase II “basket” trial at Memorial Sloan Kettering (MSK) will evaluate ado-trastuzumab emtansine across 4 cohorts of patients with HER2 amplified or HER2 overexpressed advanced lung, bladder, endometrial, and other cancers. All patients will receive ado-trastuzumab emtansine at 3.6 mg/m 2 IV every 21 days until disease progression or unacceptable toxicity. The primary endpoint is objective response rate (ORR). Patients will be molecularly selected primarily through the MSK-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT), where all patients with advanced cancers can have tumor next generation sequencing (NGS) performed with a capacity to sequence 15,000 tumors each year. MSK-IMPACT uses the Illumina HiSeq platform to screen for potentially actionable genetic alterations, including single base substitutions, indels, copy number alterations and selected fusions across 341 cancer-related genes, including HER2 amplification. HER2 amplification assessment by our NGS assay correlates well with amplification by in-situ hybridization, is less operator-dependent and is performed concurrently with the mutation profile. In the first 6 months since introducing MSK-IMPACT into routine patient care, we have already identified HER2 amplification in 7 of 227 (3%) lung cancers sequenced, 4 of 67 (6%) bladder cancers sequenced, and 2 of 50 (4%) endometrial cancers sequenced. Using a Simon optimal two-stage design, a one-sided Type I error rate α at 10% and power of 80%, a true ORR ≤ 10% will be considered unacceptable (null hypothesis) whereas a true ORR ≥ 30% will merit further study (alternative hypothesis). In each cohort, 7 patients will be accrued in the first stage; if there are no responses observed, the cohort will be closed. Otherwise, 11 additional patients will be accrued for second stage. A cohort will be deemed worthy of further investigation if ≥4 responses are observed in 18 patients. Exploratory analysis will examine the concordance among the HER2 biomarkers: gene amplification, protein overexpression and gene mutation. Citation Format: Bob T. Li, Marjorie Zauderer, Jamie Chaft, Alexander Drilon, Juliana Eng, Camelia Sima, Vicky Makker, Gopa Iyer, Yelena Janjigian, David Hyman, Maria Arcila, Jose Baselga, Mark G. Kris. Ado-trastuzumab emtansine for HER2 amplified or HER2 overexpressed cancers: A phase II “basket” trial. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr CT225. doi:10.1158/1538-7445.AM2015-CT225
Cancer Research | 2012
Anne Eaton; Camelia Sima; K. S. Panageas
Purpose: To gain FDA approval, drug companies submit evidence from clinical trials demonstrating that a drug is safe and efficacious in a certain setting. Current regulations allow oncologists to prescribe approved drugs for uses other than those for which the drug was approved (off-label use). While there seems to be a consensus that off-label use is common in oncology, there is a lack of published data on recent trends and on utilization in different populations. The strength of evidence demonstrating safety and efficacy varies for drugs used in the off-label setting, and patients may be at risk for unacceptable toxicity. The risk of toxicity may be higher among older patients, whose ability to tolerate chemotherapy is compromised by age-related organ decline and comorbidities. Our objective was to describe the prevalence of off-label use of chemotherapeutic agents in elderly breast cancer patients with Medicare coverage and to compare toxicity between patients using off-label drugs and patients using only approved drugs. Materials and Methods: We identified women 65 and older with a first primary breast cancer diagnosed in 2000–2007 from Surveillance, Epidemiology and End Results (SEER) cancer registry data linked with Medicare claims. Specific chemotherapeutic agents used following diagnosis were identified in Medicare claims and classified as on- or off-label use based on whether the drug was approved for breast cancer as of May 2012. Thirty-day toxicity (ER visit, hospitalization or death) was compared between patients whose first line regimen contained only approved drugs and patients whose first line regimen contained at least one off-label drug using the Cochran-Mantel-Haenszel test. Results: The most commonly-used approved drugs were cyclophosphamide (used in 14% of patients), doxorubicin (10%), docetaxel (5%), fluorouracil (5%), and paclitaxel (5%). In total, forty-five off-label agents were used. Vinorelbine and carboplatin were each used in 1% of patients. No other off-label drug was used in more than 109 (0.11%) of patients. Toxicity was significantly higher for patients treated with off-label agents compared to patients treated with only approved drugs, after adjusting for stage at diagnosis (p = 0.01). Conclusions: In this population-based cohort, rates of chemotherapy use were generally low, and rates of off-label use were even lower, with only 3% of patients treated off-label. Off-label use became more common as stage at diagnosis increased, with use increasing from 1% in stage 0/1 patients to 12% in stage 4 patients. The higher toxicity rates observed in patients treated with off-label drugs may indicate that these drugs are less safe than approved agents. This result will be confirmed in a matched analysis accounting for age, stage and comorbidities. Citation Information: Cancer Res 2012;72(24 Suppl):Abstract nr P3-07-09.