59 results on '"Ai Ni"'
Search Results
2. Targeting NFE2L2/KEAP1 Mutations in Advanced NSCLC With the TORC1/2 Inhibitor TAK-228
- Author
-
Paul K. Paik, Pang-Dian Fan, Besnik Qeriqi, Azadeh Namakydoust, Bobby Daly, Linda Ahn, Rachel Kim, Andrew Plodkowski, Ai Ni, Jason Chang, Rachel Fanaroff, Marc Ladanyi, Elisa de Stanchina, and Charles M. Rudin
- Subjects
Pulmonary and Respiratory Medicine ,Oncology - Abstract
Increased insight into the mutational landscape of squamous cell lung cancers (LUSCs) in the past decade has not translated into effective targeted therapies for patients with this disease. NRF2, encoded by NFE2L2, and its upstream regulator, KEAP1, control key aspects of redox balance and are frequently mutated in NSCLCs.Here, we describe the specific potent activity of TAK-228, a TORC1/2 inhibitor, in NSCLC models harboring NRF2-activating alterations and results of a phase 2 clinical trial of TAK-228 in patients with advanced NSCLC harboring NRF2-activating alterations including three cohorts (NFE2L2-mutated LUSC, KEAP1-mutated LUSC, KRAS/NFE2L2- or KEAP1-mutated NSCLC).TAK-228 was most efficacious in a LUSC cohort with NFE2L2 alterations; the overall response rate was 25% and median progression-free survival was 8.9 months. Additional data suggest that concurrent inhibition of glutaminase with the glutaminase inhibitor CB-839 might overcome metabolic resistance to therapy in these patients.TAK-228 has single-agent activity in patients with NRF2-activated LUSC. This study reframes oncogenic alterations as biologically relevant based on their downstream effects on metabolism. This trial represents, to the best of our knowledge, the first successful attempt at metabolically targeting NSCLC and identifies a promising targeted therapy for patients with LUSC, who are bereft of genotype-directed therapies.
- Published
- 2022
3. A Phase II Trial of Albumin-Bound Paclitaxel and Gemcitabine in Patients with Newly Diagnosed Stage IV Squamous Cell Lung Cancers
- Author
-
Andrew J. Plodkowski, Daniel C. McFarland, Miguel A. Villalona-Calero, Mark G. Kris, John J. Fiore, Juliana Eng, Rachel Kim, Ai Ni, Afsheen Iqbal, Charles M. Rudin, Philip Jonsson, Paul K. Paik, Mark T.A. Donoghue, Linda Ahn, and Kenneth K. Ng
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Phases of clinical research ,Pembrolizumab ,Deoxycytidine ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biomarkers, Tumor ,medicine ,Clinical endpoint ,Humans ,Neoplasms, Squamous Cell ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Taxane ,business.industry ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Gemcitabine ,Confidence interval ,Carboplatin ,Survival Rate ,Treatment Outcome ,030104 developmental biology ,Paclitaxel ,chemistry ,030220 oncology & carcinogenesis ,Female ,Patient Safety ,Albumin-Bound Paclitaxel ,business ,medicine.drug - Abstract
Purpose: Gemcitabine and albumin-bound paclitaxel (ABP) exhibit synergistic antitumor efficacy, with ABP serving to increase the intratumoral gemcitabine concentration. Both drugs are active in squamous cell lung cancers (SQCLC) and are conventional partners for carboplatin. We hypothesized that combining gemcitabine and ABP would enhance the antitumor activity in patients with advanced SQCLCs. Patients and Methods: This was a Simon two-stage, open-label, single-arm, multicenter phase II study that enrolled patients between August 1, 2015 and June 1, 2018. We enrolled 37 patients with chemotherapy-naïve, PD-L1 low/unknown advanced stage IV SQCLC. Patients were administered weekly intravenous gemcitabine (1,000 mg/m2) plus ABP (100 mg/m2) in a 3-week on, 1-week off schedule during stage I and a 2-week on, 1-week off schedule in stage II. The primary endpoint was best objective response rate (ORR). Next-generation sequencing by MSK-IMPACT was used to calculate tumor mutation burden and genome doubling and assess somatic variants for correlations with efficacy. Results: Thirty-two patients were evaluable for response assessment. The study satisfied its primary endpoint, with confirmed partial responses in 18 of 32 patients and a complete response in 1 patient [ORR 59%; 95% confidence interval (CI), 42%–74%]. Median progression-free survival (PFS), a secondary endpoint, was 7.5 (95% CI, 6.7–10.5) months. There were no unexpected toxicities. Conclusions: Gemcitabine plus ABP was a safe, tolerable, and effective first-line therapy for patients with chemotherapy-naïve SQCLCs, with an ORR and median PFS substantially higher than carboplatin doublet regimens and efficacy comparable with carboplatin plus taxane plus pembrolizumab.
- Published
- 2020
4. Harnessing Clinical Sequencing Data for Survival Stratification of Patients With Metastatic Lung Adenocarcinomas
- Author
-
David B. Solit, Kathryn C. Arbour, Matthew D. Hellmann, Mark G. Kris, Maria E. Arcila, Venkatraman E. Seshan, Axel Martin, Gregory J. Riely, Charles M. Rudin, Emmet Jordan, Ryan Ptashkin, Ronglai Shen, Marc Ladanyi, Ahmet Zehir, Arshi Arora, Michael F. Berger, and Ai Ni
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,business.industry ,Sequencing data ,Cancer ,medicine.disease ,Article ,Stratification (mathematics) ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,Prognostic model ,medicine ,business ,Routine care ,Metastatic Lung Adenocarcinoma - Abstract
PURPOSE Broad-panel sequencing of tumors facilitates routine care of people with cancer as well as clinical trial matching for novel genome-directed therapies. We sought to extend the use of broad-panel sequencing results to survival stratification and clinical outcome prediction. METHODS By using sequencing results from a cohort of 1,054 patients with advanced lung adenocarcinomas, we developed OncoCast, a machine learning tool for survival risk stratification and biomarker identification. RESULTS With OncoCast, we stratified this patient cohort into four risk groups on the basis of tumor genomic profile. Patients whose tumors harbored a high-risk profile had a median survival of 7.3 months (95% CI, 5.5 to 10.9 months) compared with a low-risk group with a median survival of 32.8 months (95% CI, 26.3 to 38.5 months) with a hazard ratio of 4.6 ( P < .001), far superior to any individual gene predictor or standard clinical characteristics. We found that comutations of both STK11 and KEAP1 are strong determinants of unfavorable prognosis with currently available therapies. In patients with targetable oncogenes (eg, EGFR, ALK, ROS1) who received targeted therapies, the tumor genetic background additionally differentiated survival with mutations in TP53 and ARID1A, which contributed to a higher risk score for shorter survival. CONCLUSION A mutational profile derived from broad-panel sequencing presents an effective genomic stratification for patient survival in advanced lung adenocarcinoma. OncoCast is available as a public resource that facilitates the incorporation of mutational data to predict individual patient prognosis and compare risk characteristics of patient populations.
- Published
- 2019
5. Incidence of infectious complications with the combination of bendamustine and an anti-CD20 monoclonal antibody
- Author
-
Brianne N. Dixon, Kristen M Sarlo, David J. Straus, and Ai Ni
- Subjects
Adult ,Bendamustine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.drug_class ,Monoclonal antibody ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,parasitic diseases ,medicine ,Retrospective analysis ,Bendamustine Hydrochloride ,Humans ,Anti cd20 ,Retrospective Studies ,business.industry ,Incidence ,Lymphoma, Non-Hodgkin ,Incidence (epidemiology) ,Antibodies, Monoclonal ,Hematology ,humanities ,body regions ,030220 oncology & carcinogenesis ,Rituximab ,business ,030215 immunology ,medicine.drug - Abstract
Combination of bendamustine (B) and rituximab (R) has been associated with opportunistic infections (OI) in case reports. This retrospective analysis evaluated the incidence, risk factors, and types of infectious complications (IC) in adults with CD20+ non-Hodgkin lymphoma who received ≥2 cycles of B and either R or ofatumumab. Infection data were collected up to 1-year post-B-based treatment. Potential risk factors for IC were assessed using univariate analysis with Fisher's exact test. Four-hundred and sixteen patients were included. Incidence of IC and OI was 20 and 6%, respectively. Viral (
- Published
- 2019
6. The survival benefit of anti-HER2 treatment in the management of small (T1mic, T1a, T1b, T1c), node-negative HER2+ breast cancer
- Author
-
Kai Conrad Cecil Johnson, Ai Ni, Michael Grimm, Sagar D. Sardesai, Daniel G. Stover, Mathew Amprayil Cherian, Margaret Elena Gatti-Mays, Ashley Pariser, Preeti Khetarpal Sudheendra, Nicole Olivia Williams, Jeffrey Bryan VanDeusen, Bhuvaneswari Ramaswamy, and Robert Wesolowski
- Subjects
Cancer Research ,Oncology - Abstract
532 Background: Limited compelling prospective and retrospective data regarding the added benefit of anti-HER2 therapy in the management of small, node-negative HER2-positive breast cancer (HER2+BC) exists, in part due to differences in outcome reporting, unmatched analyses, and a lack of head-to-head comparisons. As a result, national guideline committees find themselves unable to confidently recommend anti-HER2 therapy and clinicians are left to exercise clinical judgement on whether the use of anti-HER2 therapy should be considered for such patients. Methods: Our team performed a multi-institutional retrospective analysis using the ASCO CancerLinQ database, with a focus on clinical data from small, node-negative HER2+BC patients diagnosed between 2010 to 2021. We compared clinical outcomes between those who received adjuvant trastuzumab therapy, with or without chemotherapy, to those who did not, with our primary outcomes being invasive disease-free survival (iDFS) and overall survival (OS). We performed both a univariate and multivariate analysis, using a Cox proportional hazard model to control for factors including age, ethnicity, body mass index, hormone status, tumor grade, histology type, BRCA status, region, and smoking history. Additionally, a three-arm univariate analysis was performed comparing untreated patients to trastuzumab alone versus combination therapy. Results: In total, 1206 patients met inclusion criteria, including 779 patients who received trastuzumab with or without chemotherapy. We found a statistically significant improvement in both iDFS (HR 0.73, p = 0.01) and OS (HR 0.63, p = 0.027) on univariate analysis for those receiving anti-HER2 therapy. Similarly on multivariate analysis, iDFS (HR 0.75, p = 0.030) and OS (HR 0.61, p = 0.029) were improved in those who received therapy, regardless of tumor size. Our three-arm univariate analysis involving no treatment (n = 427), trastuzumab monotherapy (n = 169), and combination therapy (n = 578) found that iDFS was significantly improved for both treatment arms compared to observation alone (p = 0.006), whereas OS trended towards significance in the treatment arms but did not reach this target (p = 0.061). No significant difference was noted between treatment arms. Conclusions: Our analysis found a statistically significant improvement in iDFS and OS when patients with small, node negative, HER2+BC received adjuvant anti-HER2 therapy with or without chemotherapy as compared to observation. From our univariate three-arm comparison, it appears that trastuzumab provides the majority of benefit to patients in terms of DFS, but this result is exploratory. Further investigation is warranted, including meta-analyses to better characterize the degree of benefit seen with anti-HER2 treatment. For now, this data adds to evidence suggesting added benefit with therapy over observation.
- Published
- 2022
7. Follicular lymphoma in the modern era: survival, treatment outcomes, and identification of high-risk subgroups
- Author
-
Anna Alperovich, Steven M. Horwitz, Jacob D. Soumerai, Philip Caron, Venkatraman E. Seshan, Anas Younes, Paul A. Hamlin, Connie Lee Batlevi, Lorenzo Falchi, Anita Kumar, Ai Ni, Alison J. Moskowitz, Audrey Hamilton, Colette Owens, Matthew J. Matasar, Craig H. Moskowitz, Ariela Noy, Zhitao Ying, Katy Smith, Erel Joffe, David J. Straus, Andrew D. Zelenetz, Fushen Sha, Lia Palomba, and Gottfried von Keudell
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Disease-free survival ,Follicular lymphoma ,Risk Assessment ,lcsh:RC254-282 ,Article ,Young Adult ,Targeted therapies ,Medical research ,International Prognostic Index ,Risk Factors ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Public Health Surveillance ,Young adult ,Lymphoma, Follicular ,Survival rate ,Survival analysis ,Aged ,Neoplasm Staging ,Retrospective Studies ,Aged, 80 and over ,B-cell lymphoma ,business.industry ,Disease Management ,Retrospective cohort study ,Hematology ,Middle Aged ,Prognosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Combined Modality Therapy ,Survival Analysis ,Lymphoma ,Survival Rate ,Treatment Outcome ,Female ,Neoplasm Grading ,business ,Follow-Up Studies - Abstract
Patients with follicular lymphoma (FL) frequently require multiple treatments during their disease course; however, survival based on lines of treatment remains poorly described in the post-rituximab era. Also, the Follicular Lymphoma International Prognostic Index (FLIPI) score was developed to predict survival at diagnosis, yet it remains unknown whether increase in FLIPI score following an initial observation period is associated with less-favorable outcomes. To address these knowledge gaps, we retrospectively studied 1088 patients with FL grade 1–3A managed between 1998 and 2009 at our institution. Median overall survival (OS) and progression-free survival (PFS) after first-line treatment were not reached and 4.73 years, respectively. Following successive lines of treatment, years of median OS and PFS were, respectively: after second-line, 11.7 and 1.5; third-line, 8.8 and 1.1; fourth-line, 5.3 and 0.9; fifth-line, 3.1 and 0.6; sixth-line, 1.9 and 0.5. In initially observed, subsequently treated patients, FLIPI score increase after observation was associated with inferior survival following first-line treatment. The reduced survival we observed after second-line and later therapy supports the development of new treatments for relapsed patients and benchmarks historical targets for clinical endpoints. This study also highlights the utility of changes in FLIPI score at diagnosis and after observation in identifying patients likely to have worse outcomes.
- Published
- 2020
8. Evaluation of the CLL-IPI in relapsed and refractory chronic lymphocytic leukemia in idelalisib phase-3 trials
- Author
-
Andrew D. Zelenetz, Ronald L. Dubowy, Jeffrey A. Jones, Jacob D. Soumerai, Richard R. Furman, Ai Ni, Jeffrey P. Sharman, Julie Huang, Michael Hallek, Lyndah Dreiling, Adeboye H. Adewoye, and Guan Xing
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Chronic lymphocytic leukemia ,Immunoglobulin Variable Region ,Kaplan-Meier Estimate ,Relapsed CLL ,Models, Biological ,Risk Assessment ,Article ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Multicenter Studies as Topic ,In patient ,neoplasms ,Aged ,Quinazolinones ,Aged, 80 and over ,business.industry ,Hematology ,Middle Aged ,Prognosis ,medicine.disease ,Leukemia, Lymphocytic, Chronic, B-Cell ,Lymphoma ,Leukemia ,Treatment Outcome ,Clinical Trials, Phase III as Topic ,Drug Resistance, Neoplasm ,Purines ,030220 oncology & carcinogenesis ,Prognostic model ,Female ,Neoplasm Recurrence, Local ,Refractory Chronic Lymphocytic Leukemia ,Immunoglobulin Heavy Chains ,business ,Idelalisib ,Follow-Up Studies ,030215 immunology - Abstract
The CLL-IPI is a risk-weighted prognostic model for previously untreated patients with chronic lymphocytic leukemia (CLL), but has not been evaluated in patients with relapsed CLL or on novel therapies. We evaluated the CLL-IPI in 897 patients with relapsed/refractory CLL in 3 randomized trials testing idelalisib (PI3Kδ inhibitor). The CLL-IPI identified patients as low (2.2%), intermediate (12.8%), high (48.7%), and very high (36.2%) risk, and was prognostic for survival (log-rank p65, β2-microglobulin >3.5mg/L, unmutated immunoglobulin heavy chain variable region gene, and deletion 17p/TP53 mutation were independently prognostic, but Rai I-IV or Binet B/C was not. The CLL-IPI is prognostic for survival in relapsed CLL and with idelalisib therapy. However, low/intermediate risk is uncommon, and regression parameters of individual factors in this risk-weighted model appear different in relapsed CLL. Reassessment of the weighting of the individual variables might optimize the model in this setting.
- Published
- 2018
9. Frequency of Brain Metastases and Multikinase Inhibitor Outcomes in Patients With RET–Rearranged Lung Cancers
- Author
-
Vamsidhar Velcheti, Tony Mok, Thomas Filleron, Nir Peled, Ai Ni, Jürgen Wolf, Julie Milia, Dwight H. Owen, Jessica J. Lin, Bob T. Li, Benjamin Besse, Justin F. Gainor, Isabella Bergagnini, David P. Carbone, M. Offin, Julien Mazieres, Alexander Drilon, D. Ross Camidge, Mark M. Awad, Vaios Hatzoglou, Gregory J. Riely, Oliver Gautschi, and Mark G. Kris
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Alectinib ,medicine.medical_specialty ,Everolimus ,Cabozantinib ,Sunitinib ,business.industry ,Vandetanib ,medicine.disease ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Cumulative incidence ,business ,Lung cancer ,medicine.drug ,Brain metastasis - Abstract
Introduction In ret proto-oncogene (RET)–rearranged lung cancers, data on the frequency of brain metastases and, in particular, the outcomes of multikinase inhibitor therapy in patients with intracranial disease are not well characterized. Methods A global, multi-institutional registry (cohort A, n = 114) and a bi-institutional data set (cohort B, n = 71) of RET-rearranged lung cancer patients were analyzed. Patients were eligible if they had stage IV lung cancers harboring a RET rearrangement by local testing. The incidence of brain metastases and outcomes with multikinase inhibitor therapy were determined. Results The frequency of brain metastases at the time of diagnosis of stage IV disease was 25% (95% confidence interval [CI]: 18%–32%) in all patients from both cohorts. The lifetime prevalence of brain metastasis in stage IV disease was 46% (95% CI: 34%–58%) in patients for whom longitudinal data was available. The cumulative incidence of brain metastases was significantly different (p = 0.0039) between RET-, ROS1-, and ALK receptor tyrosine kinase (ALK)–rearranged lung cancers, with RET intermediate between the other two groups. Although intracranial response data was not available in cohort A, the median progression-free survival of multikinase inhibitor therapy (cabozantinib, vandetanib, or sunitinib) in patients with brain metastases was 2.1 months (95% CI: 1.3–2.9 months, n = 10). In cohort B, an intracranial response was observed in 2 of 11 patients (18%) treated with cabozantinib, vandetanib (± everolimus), ponatinib, or alectinib; the median overall progression-free survival (intracranial and extracranial) was 3.9 months (95% CI: 2.0–4.9 months). Conclusions Brain metastases occur frequently in RET-rearranged lung cancers, and outcomes with multikinase inhibitor therapy in general are suboptimal. Novel RET-directed targeted therapy strategies are needed.
- Published
- 2018
10. PD-L1 expression, tumor mutational burden, and response to immunotherapy in patients with MET exon 14 altered lung cancers
- Author
-
Mizuki Nishino, Andrew J. Plodkowski, Joseph Montecalvo, Chebli Mrad, Charles M. Rudin, Lynette M. Sholl, Wei-Chu Victoria Lai, Mark G. Kris, Giulia Costanza Leonardi, Ai Ni, Renato Umeton, Mark M. Awad, Joshua K. Sabari, N. Rekhtman, Michael Offin, A. Drilon, Bob T. Li, Ruqin Chen, Darragh Halpenny, Catherine A. Shu, Kathryn C. Arbour, Matthew D. Hellmann, G. J. Riely, Isabella Bergagnini, Jules L. Dienstag, and Paul K. Paik
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Adenocarcinoma ,B7-H1 Antigen ,Cohort Studies ,03 medical and health sciences ,Exon ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Stage (cooking) ,Aged ,Aged, 80 and over ,Lung ,business.industry ,High-Throughput Nucleotide Sequencing ,Original Articles ,Exons ,Hematology ,Immunotherapy ,Middle Aged ,Proto-Oncogene Proteins c-met ,Prognosis ,MET Exon 14 Skipping Mutation ,Immune checkpoint ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cohort ,Immunohistochemistry ,Female ,business ,Follow-Up Studies - Abstract
BACKGROUND: MET exon 14 alterations are actionable oncogenic drivers. Durable responses to MET inhibitors are observed in patients with advanced MET exon 14-altered lung cancers in prospective trials. In contrast, the activity of immunotherapy, PD-L1 expression and tumor mutational burden (TMB) of these tumors and are not well characterized. PATIENTS AND METHODS: Patients with MET exon 14-altered lung cancers of any stage treated at two academic institutions were identified. A review of clinicopathologic and molecular features, and an analysis of response to single-agent or combination immune checkpoint inhibition were conducted. PD-L1 immunohistochemistry was carried out and TMB was calculated by estimation from targeted next-generation sequencing panels. RESULTS: We identified 147 patients with MET exon 14-altered lung cancers. PD-L1 expression of 0%, 1%–49%, and ≥50% were 37%, 22%, and 41%, respectively, in 111 evaluable tumor samples. The median TMB of MET exon 14-altered lung cancers was lower than that of unselected non-small-cell lung cancers (NSCLCs) in both independently evaluated cohorts: 3.8 versus 5.7 mutations/megabase (P
- Published
- 2018
11. Concurrent Alterations in EGFR-Mutant Lung Cancers Associated with Resistance to EGFR Kinase Inhibitors and Characterization of MTOR as a Mediator of Resistance
- Author
-
Gregory J. Riely, Marc Ladanyi, Ryan Kim, Ken Suzawa, Matthew D. Hellmann, Helena A. Yu, Maria E. Arcila, Ahmet Zehir, Ai Ni, Bob T. Li, Michael F. Berger, Romel Somwar, Emmet Jordan, Mark G. Kris, and David B. Solit
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Antineoplastic Agents ,Drug resistance ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,Germline mutation ,Cell Line, Tumor ,Biomarkers, Tumor ,medicine ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Mutation ,business.industry ,Kinase ,TOR Serine-Threonine Kinases ,Cancer ,Genomics ,Prognosis ,medicine.disease ,ErbB Receptors ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Erlotinib ,business ,medicine.drug - Abstract
Purpose: To identify molecular factors that determine duration of response to EGFR tyrosine kinase inhibitors and to identify novel mechanisms of drug resistance, we molecularly profiled EGFR-mutant tumors prior to treatment and after progression on EGFR TKI using targeted next-generation sequencing. Experimental Design: Targeted next-generation sequencing was performed on 374 consecutive patients with metastatic EGFR-mutant lung cancer. Clinical data were collected and correlated with somatic mutation data. Erlotinib resistance due to acquired MTOR mutation was functionally evaluated by in vivo and in vitro studies. Results: In 200 EGFR-mutant pretreatment samples, the most frequent concurrent alterations were mutations in TP53, PIK3CA, CTNNB1, and RB1 and focal amplifications in EGFR, TTF1, MDM2, CDK4, and FOXA1. Shorter time to progression on EGFR TKI was associated with amplification of ERBB2 (HR = 2.4, P = 0.015) or MET (HR = 3.7, P = 0.019), or mutation in TP53 (HR = 1.7, P = 0.006). In the 136 posttreatment samples, we identified known mechanisms of acquired resistance: EGFR T790M (51%), MET (7%), and ERBB2 amplifications (5%). In the 38 paired samples, novel acquired alterations representing putative resistance mechanisms included BRAF fusion, FGFR3 fusion, YES1 amplification, KEAP1 loss, and an MTOR E2419K mutation. Functional studies confirmed the contribution of the latter to reduced sensitivity to EGFR TKI in vitro and in vivo. Conclusions: EGFR-mutant lung cancers harbor a spectrum of concurrent alterations that have prognostic and predictive significance. By utilizing paired samples, we identified several novel acquired alterations that may be relevant in mediating resistance, including an activating mutation in MTOR further validated functionally. Clin Cancer Res; 24(13); 3108–18. ©2018 AACR.
- Published
- 2018
12. Molecular Determinants of Response to Anti–Programmed Cell Death (PD)-1 and Anti–Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non–Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing
- Author
-
Andrew J. Plodkowski, Darragh Halpenny, Jennifer L. Sauter, Barry S. Taylor, Ronglai Shen, Walid K. Chatila, Kurt A. Schalper, Kathryn C. Arbour, Matthew D. Hellmann, Ai Ni, Marc Ladanyi, Jedd D. Wolchok, Taha Merghoub, Philip Jonsson, Justin F. Gainor, Travis J. Hollmann, Natasha Rekhtman, David B. Solit, Konnor La, Mark G. Kris, Nikolaus Schultz, Michael F. Berger, Nicholas D. Socci, Jamie E. Chaft, Ahmet Zehir, Hira Rizvi, Alexandra Snyder, Gregory J. Riely, Niamh Long, Maria E. Arcila, Charles M. Rudin, and Francisco Sanchez-Vega
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Hazard ratio ,STK11 ,ORIGINAL REPORTS ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Internal medicine ,PD-L1 ,medicine ,Carcinoma ,biology.protein ,Lung cancer ,business ,Exome sequencing - Abstract
Purpose Treatment of advanced non–small-cell lung cancer with immune checkpoint inhibitors (ICIs) is characterized by durable responses and improved survival in a subset of patients. Clinically available tools to optimize use of ICIs and understand the molecular determinants of response are needed. Targeted next-generation sequencing (NGS) is increasingly routine, but its role in identifying predictors of response to ICIs is not known. Methods Detailed clinical annotation and response data were collected for patients with advanced non–small-cell lung cancer treated with anti–programmed death-1 or anti–programmed death-ligand 1 [anti-programmed cell death (PD)-1] therapy and profiled by targeted NGS (MSK-IMPACT; n = 240). Efficacy was assessed by Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1, and durable clinical benefit (DCB) was defined as partial response/stable disease that lasted > 6 months. Tumor mutation burden (TMB), fraction of copy number–altered genome, and gene alterations were compared among patients with DCB and no durable benefit (NDB). Whole-exome sequencing (WES) was performed for 49 patients to compare quantification of TMB by targeted NGS versus WES. Results Estimates of TMB by targeted NGS correlated well with WES (ρ = 0.86; P < .001). TMB was greater in patients with DCB than with NDB ( P = .006). DCB was more common, and progression-free survival was longer in patients at increasing thresholds above versus below the 50th percentile of TMB (38.6% v 25.1%; P < .001; hazard ratio, 1.38; P = .024). The fraction of copy number–altered genome was highest in those with NDB. Variants in EGFR and STK11 associated with a lack of benefit. TMB and PD-L1 expression were independent variables, and a composite of TMB plus PD-L1 further enriched for benefit to ICIs. Conclusion Targeted NGS accurately estimates TMB and elevated TMB further improved likelihood of benefit to ICIs. TMB did not correlate with PD-L1 expression; both variables had similar predictive capacity. The incorporation of both TMB and PD-L1 expression into multivariable predictive models should result in greater predictive power.
- Published
- 2018
13. Zanubrutinib, Obinutuzumab, and Venetoclax in Chronic Lymphocytic Leukemia: Early MRD Kinetics Define a High-Risk Patient Cohort with Delayed Bone Marrow Undetectable MRD and Earlier Post-Treatment MRD Recurrence
- Author
-
Jeffrey A. Barnes, Jenny Wu, Anita A Kumar, Krista J Scorsune, Allison P. Jacob, Jade Ruiters, Connie Lee Batlevi, Rosalba Martignetti, Chaya Friedman, Venkatraman E. Seshan, Daneal Portman, Audrey Hamilton, Ai Ni, Anthony R. Mato, Morgan Choma, Meghan C. Thompson, Clare Grieve, Jane Huang, Elizabeth Simkins, Ahmet Dogan, Tak Takvorian, Jason Carter, Brianne McGree, Colette Owens, Julia M Lynch, Ariela Noy, Puja Chadha, Sidney Sechio, Mikhail Roshal, Erel Joffe, Joanna Mi, M. Lia Palomba, Kelsey Flaherty, Lindsey E. Roeker, Natascha Nolet, Andrew D. Zelenetz, Stephanie Hughes, P. Connor Johnson, Matthew J. Matasar, Omar Abdel-Wahab, Jacob D. Soumerai, Juliana M.L. Biondo, Neena Mahajan, Ephraim P. Hochberg, and Jeremy S. Abramson
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Venetoclax ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Obinutuzumab ,Internal medicine ,Cohort ,medicine ,Bone marrow ,Post treatment ,business - Abstract
Background: Venetoclax (Ven; BH3 mimetic) and Obinutuzumab (O; CD20 antibody) is an approved, fixed-duration regimen (one year) that induces undetectable minimum residual disease (uMRD) and durable remissions in treatment naïve patients (pts) with chronic lymphocytic leukemia (CLL; Fischer NEJM 2019). In the CLARITY trial of venetoclax-ibrutinib (BTK inhibitor; BTKi) in relapsed or refractory CLL, peripheral blood (PB) MRD response kinetics predicted bone marrow (BM) uMRD and were associated with progression-free survival (Rawstron EHA 2020). We explored MRD as a biomarker to direct treatment duration in an investigator-initiated phase 2 trial of Zanubrutinib (BTKi) and O-Ven (BOVen). We hypothesize that early MRD kinetics will identify a defined cohort of pts with delayed BM MRD clearance, and early recurrent detectable MRD after discontinuation of treatment in pts attaining uMRD. Methods: In this multicenter, phase 2 trial (NCT03824483), eligible pts had previously untreated CLL requiring treatment (iwCLL), ECOG PS ≤2, absolute neutrophil count (ANC) ≥1,000/ul, platelets (PLT) ≥75,000/ul (ANC ≥0/ul, PLT ≥20,000/ul if due to CLL). Informed consent was obtained from all pts. BOVen was administered in 28-day (D) cycles (C): Zanubrutinib 160 mg by mouth (PO) twice daily starting D1; Obinutuzumab 1000 mg IV D1 (split D1-2 if lymphocyte count ≥25,000/ul or lymph node ≥5cm), D8, D15 of C1, and D1 of C2-8; Venetoclax ramp up initiated C3D1 (target 400 mg PO daily). MRD was evaluated by flow cytometry (MRD-FC) and immunosequencing (MRD-IS; Adaptive ClonoSEQ) with uMRD defined as ≤10 -4 for flow and ≤10 -5 for IS. Treatment consisted of 8-24 cycles with duration determined by prespecified MRD criteria. Beginning on C7D1 then every 2 cycles, pts with PB uMRD-FC had BM within 14 days. If BM uMRD, PB MRD was repeated after two additional cycles. Pts with confirmed uMRD-FC in PB and BM discontinued therapy and entered posttreatment surveillance. Response was assessed per iwCLL. Adverse events (AE) were assessed per CTCAE v5. MRD-IS failure free survival (FFS) was calculated from end-of-treatment (EOT) to the date of detectable MRD-IS (≥10 -5) using the Kaplan-Meier method. Results: The study accrued 39 pts (03/19-10/19): median age 59 years (23-73), 3:1 male predominance, 28/39 (72%) IGHV unmutated, 5/39 (12.8%) del(17p)/TP53M. All pts were evaluable for toxicity with 37 evaluable for efficacy. At a median follow up of 26+ months (mo; 4.5-30.5+), 95% (35/37) pts achieved uMRD-FC in PB, among whom 33 (94%) also achieved uMRD-IS. BM uMRD-FC was seen in 89% (33/37) at a median time of 8 mo (6-16), all of whom met prespecified MRD criteria and discontinued therapy after a median of 10 mo (8-18). Three pts discontinued therapy with persistent detectable BM MRD after 24 cycles. The most common AEs were neutropenia (51%), thrombocytopenia (44%), diarrhea (44%), infusion related reaction (41%) and bruising (41%). The most common grade ≥3 AE was neutropenia (15%). No laboratory or clinical TLS occurred (Howard definition). A ≥400-fold reduction in PB MRD-IS after 4 cycles (ΔMRD400) was selected using the Youden Index and was highly predictive of attaining BM uMRD in ≤8mo (sensitivity 88% [21/24], specificity 100% [11/11], PPV 100% [21/21], NPV 79% [11/14]. As a result, the median duration of therapy was shorter among patients who achieved ΔMRD400 (8 vs 13 mo). Of 33 pts who met prespecified uMRD criteria and stopped therapy, 31 (94%) remain uMRD-FC following a median 15 mo (0-20) from EOT, and 2 pts had recurrent MRD (1 with PD recaptured PB uMRD with retreatment). Of 33 pts who discontinued therapy after achieving the prespecified MRD endpoint, MRD-IS was evaluated every 3 months in 31 pts for a median of 12 mo (range, 3-18) from EOT. MRD-IS FFS was longer in pts who achieved ΔMRD400 (log rank p Conclusion: BOVen achieved frequent, durable uMRD. All pts completed therapy (median 10 mo treatment), including 89% (33/37) who met the prespecified PB/BM uMRD endpoint. With a median posttreatment follow-up of 15 mo, 31 (94%) remain uMRD-FC. ΔMRD400 identified a cohort of pts (40%) with delayed BM MRD clearance and earlier MRD recurrence, despite longer treatment duration. ΔMRD400 warrants further study as a predictive biomarker for treatment duration. Figure 1 Figure 1. Disclosures Soumerai: Seattle Genetics: Consultancy; AstraZeneca: Consultancy; BeiGene: Consultancy, Research Funding; BMS: Consultancy; Adaptive Biotechnologies: Consultancy, Research Funding; AbbVie: Consultancy; TG Therapeutics: Consultancy, Research Funding; BostonGene: Research Funding; GlaxoSmithKline: Research Funding. Mato: Janssen: Consultancy, Research Funding; LOXO: Consultancy, Research Funding; Johnson and Johnson: Consultancy, Research Funding; Acerta/AstraZeneca: Consultancy, Research Funding; DTRM BioPharma: Consultancy, Research Funding; Genmab: Research Funding; AstraZeneca: Consultancy; MSKCC: Current Employment; Genentech: Consultancy, Research Funding; Sunesis: Consultancy, Research Funding; Nurix: Research Funding; AbbVie: Consultancy, Research Funding; Adaptive Biotechnologies: Consultancy, Research Funding; Pharmacyclics LLC, an AbbVie Company: Consultancy, Research Funding; TG Therapeutics: Consultancy, Other: DSMB, Research Funding; BeiGene: Consultancy, Research Funding. Dogan: Seattle Genetics: Consultancy; Peer View: Honoraria; Takeda: Consultancy, Research Funding; Roche: Consultancy, Research Funding; Physicians' Education Resource: Honoraria; EUSA Pharma: Consultancy. Joffe: Epizyme: Consultancy; AstraZeneca: Consultancy. Hochberg: Leuko: Consultancy; Intervention Insights: Consultancy. Abramson: Bluebird Bio: Consultancy; Morphosys: Consultancy; Bristol-Myers Squibb Company: Consultancy, Research Funding; Kymera: Consultancy; BeiGene: Consultancy; Novartis: Consultancy; C4 Therapeutics: Consultancy; Genmab: Consultancy; EMD Serono: Consultancy; Kite Pharma: Consultancy; Incyte Corporation: Consultancy; Astra-Zeneca: Consultancy; Allogene Therapeutics: Consultancy; Seagen Inc.: Research Funding; AbbVie: Consultancy; Karyopharm: Consultancy; Genentech: Consultancy. Batlevi: TouchIME: Honoraria; BMS: Current holder of individual stocks in a privately-held company; Medscape: Honoraria; GLG Pharma: Consultancy; Dava Oncology: Honoraria; Kite Pharma: Consultancy; Juno/Celgene: Consultancy; ADC Therapeutics: Consultancy; Life Sciences: Consultancy; Moderna: Current holder of individual stocks in a privately-held company; Regeneron: Current holder of individual stocks in a privately-held company; Viatris: Current holder of individual stocks in a privately-held company; Pfizer: Current holder of individual stocks in a privately-held company; Karyopharm: Consultancy; TG Therapeutics: Consultancy; Memorial Sloan Kettering Cancer Center: Current Employment; Seattle Genetics: Consultancy; Bayer: Research Funding; Xynomic: Research Funding; Roche/Genentech: Research Funding; Novartis: Research Funding; Epizyme: Research Funding; Janssen: Research Funding; Autolus: Research Funding. Matasar: Rocket Medical: Consultancy, Research Funding; Merck Sharp & Dohme: Current holder of individual stocks in a privately-held company; Juno Therapeutics: Consultancy; Merck: Consultancy; Genentech, Inc.: Consultancy, Honoraria, Research Funding; Bayer: Consultancy, Honoraria, Research Funding; IGM Biosciences: Research Funding; GlaxoSmithKline: Honoraria, Research Funding; F. Hoffmann-La Roche Ltd: Consultancy, Honoraria, Research Funding; Memorial Sloan Kettering Cancer Center: Current Employment; Teva: Consultancy; TG Therapeutics: Consultancy, Honoraria; Pharmacyclics: Honoraria, Research Funding; Janssen: Honoraria, Research Funding; Takeda: Consultancy, Honoraria; Seattle Genetics: Consultancy, Honoraria, Research Funding; ImmunoVaccine Technologies: Consultancy, Honoraria, Research Funding; Daiichi Sankyo: Consultancy. Noy: Rafael Parhma: Research Funding; Morphosys: Consultancy; Targeted Oncology: Consultancy; Medscape: Consultancy; Pharmacyclics: Consultancy, Research Funding; Janssen: Consultancy, Honoraria; Epizyme: Consultancy. Palomba: Ceramedix: Honoraria; Seres: Honoraria, Other: Stock, Patents & Royalties, Research Funding; Notch: Honoraria, Other: Stock; Novartis: Consultancy; Kite: Consultancy; PCYC: Consultancy; BeiGene: Consultancy; Lygenesis: Honoraria; Nektar: Honoraria; Juno: Patents & Royalties; Wolters Kluwer: Patents & Royalties; Rheos: Honoraria; Magenta: Honoraria; Pluto: Honoraria; WindMIL: Honoraria; Priothera: Honoraria. Kumar: Abbvie Pharmaceuticals: Research Funding; Pharmacyclics: Research Funding; Kite Pharmaceuticals: Other: advisory board , Research Funding; Celgene: Honoraria, Other: advisory board, Research Funding; Astra Zeneca: Honoraria, Other: Advisory Board, Research Funding; Adaptive Biotechnologies, Celgene, Abbvie Pharmaceticals, Pharmacyclics, Seattle Genetics: Research Funding; Seattle Genetics: Research Funding. Roeker: AbbVie, AstraZeneca, Janssen, LOXO, Pharmacyclics, TG Therapeutics, Vaniam Group, Verastem: Consultancy; Pfizer: Consultancy, Research Funding; Pharmacyclics: Consultancy; TG Therapeutics: Consultancy; Loxo Oncology: Consultancy; Abbot Laboratories: Current equity holder in publicly-traded company. Thompson: VJHemOnc: Honoraria; MJH Life Sciences: Honoraria; Curio Science: Honoraria. Roshal: Physicians' Education Resource: Other: Provision of services; Auron Therapeutics: Other: Ownership / Equity interests; Provision of services; Celgene: Other: Provision of services. Huang: BeiGene: Current Employment, Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company, Divested equity in a private or publicly-traded company in the past 24 months, Other: Travel, Accommodations, Expenses; Protara Therapeutics: Current holder of individual stocks in a privately-held company, Membership on an entity's Board of Directors or advisory committees, Other: TRAVEL, ACCOMMODATIONS, EXPENSES (paid by any for-profit health care company). Biondo: Roche: Current holder of individual stocks in a privately-held company; Genentech, Inc.: Current Employment. Wu: Genentech, Inc.: Current Employment; Roche/GNE: Current equity holder in publicly-traded company, Current holder of individual stocks in a privately-held company, Current holder of stock options in a privately-held company. Jacob: Adaptive Biotechnologies: Current Employment. Abdel-Wahab: H3B Biomedicine: Consultancy, Research Funding; Foundation Medicine Inc: Consultancy; Merck: Consultancy; Prelude Therapeutics: Consultancy; LOXO Oncology: Consultancy, Research Funding; Lilly: Consultancy; AIChemy: Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees; Envisagenics Inc.: Current holder of stock options in a privately-held company, Membership on an entity's Board of Directors or advisory committees. Zelenetz: Novartis: Honoraria; Genentech/Roche: Honoraria, Research Funding; BMS/Celgene/JUNO: Honoraria, Other; AstraZeneca: Honoraria; MethylGene: Research Funding; Pharmacyclics: Honoraria; Amgen: Honoraria; MEI Pharma: Honoraria, Research Funding; Gilead: Honoraria, Research Funding; Verastem: Honoraria; Beigene: Honoraria, Other, Research Funding; Abbvie: Honoraria, Research Funding; SecuraBio: Honoraria; Janssen: Honoraria; Gilead: Honoraria; MorphoSys: Honoraria; NCCN: Other; LFR: Other. OffLabel Disclosure: Zanubrutinib is administered off-label in combination with venetoclax and obinutuzumab for patients with CLL/SLL.
- Published
- 2021
14. Twice weekly pulse and daily continuous-dose erlotinib as initial treatment for patients with epidermal growth factor receptor-mutant lung cancers and brain metastases
- Author
-
William Pao, Helena A. Yu, Robert J. Young, Ai Ni, Mark G. Kris, Linda Ahn, Christopher R. Rodriguez, Gregory J. Riely, Kathryn Beal, Sara A. Hayes, Mariza Daras, and Kathryn C. Arbour
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Nausea ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Medicine ,Epidermal growth factor receptor ,Adverse effect ,biology ,business.industry ,Cancer ,medicine.disease ,Rash ,Regimen ,030104 developmental biology ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,biology.protein ,Erlotinib ,medicine.symptom ,business ,medicine.drug - Abstract
BACKGROUND In a phase 1 study of pulse/continuous-dose erlotinib, no patient had disease progression in the central nervous system (CNS). This expansion cohort of the phase 1 study tested the same regimen in a cohort of individuals with epidermal growth factor receptor (EGFR)–mutant lung cancers with untreated brain metastases. METHODS Patients had not received EGFR tyrosine kinase inhibitors or radiation for brain metastases. All received 1200 mg of erlotinib on days 1 and 2 and 50 mg on days 3 to 7 weekly. The primary endpoints were the overall and CNS response rates (according to version 1.1 of the Response Evaluation Criteria in Solid Tumors). RESULTS Between May 2015 and August 2016, 19 patients were enrolled. Forty-two percent of the patients had target brain lesions, and the median size of the target brain lesions was 13 mm. Overall, 14 patients (74%; 95% confidence interval [CI], 51%-89%) had partial responses. The response rate in brain metastases was 75%. The overall median progression-free survival was 10 months (95% CI, 7 months to not reached). Only 3 patients (16%) had CNS progression. To date, 4 patients required CNS radiation at some time during their course. The adverse events (any grade) seen in 10% or more of the patients were rash, diarrhea, nausea, an increase in alanine aminotransferase, and fatigue. CONCLUSIONS Pulse/continuous-dose erlotinib produced a 74% overall response rate and a 75% response rate in brain metastases in patients with EGFR-mutant lung cancers and untreated brain metastases. CNS control persisted even after progression elsewhere. Although this regimen did not improve progression-free survival or delay the emergence of EGFR T790M, it prevented progression in the brain and could be useful in situations in which CNS control is critical. Cancer 2017. © 2017 American Cancer Society.
- Published
- 2017
15. Antibody-mediated thyroid dysfunction during T-cell checkpoint blockade in patients with non-small-cell lung cancer
- Author
-
L. Cambridge, M.K. Kasler, Stephanie Fish, Dennis Stephens, Carlos J. Rodriguez, Hira Rizvi, R. Pollina, Jamie E. Chaft, Juan C. Osorio, Taha Merghoub, Matthew D. Hellmann, Jedd D. Wolchok, Charles M. Rudin, and Ai Ni
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,endocrine system ,medicine.medical_specialty ,Adolescent ,Drug-Related Side Effects and Adverse Reactions ,endocrine system diseases ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Thyroid Gland ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,Hyperthyroidism ,Thyroid function tests ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Humans ,Medicine ,Adverse effect ,Lung cancer ,Aged ,Neoplasm Staging ,biology ,medicine.diagnostic_test ,business.industry ,Thyroid ,Antibodies, Monoclonal ,Cancer ,Original Articles ,Hematology ,Middle Aged ,medicine.disease ,Blockade ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Female ,Antibody ,business - Abstract
Programmed cell death protein-1 (PD-1) blockade therapies have demonstrated durable responses and prolonged survival in a variety of malignancies. Treatment is generally well tolerated although immune-related adverse events (irAEs) can occur. Autoimmune thyroid dysfunction is among the most common irAE, but an assessment of the clinical, mechanistic, and immunologic features has not been previously described.Patients with advanced non-small-cell lung cancer (NSCLC) treated with pembrolizumab at Memorial Sloan Kettering Cancer Center (n = 51) as part of KEYNOTE-001 (NCT01295827) were included. Thyroid function test and anti-thyroid antibodies were assessed prospectively at each study visit, beginning before the first treatment. Frequency of development of thyroid dysfunction, association with anti-thyroid antibodies, clinical course, and relationship with progression-free survival and overall survival to treatment with pembrolizumab was evaluated.Of 51 patients treated, 3 were hypothyroid and 48 were not at baseline. Ten of 48 [21%, 95% confidence interval (CI) 10% to 35%] patients developed thyroid dysfunction requiring thyroid replacement. Anti-thyroid antibodies were present in 8 of 10 patients who developed thyroid dysfunction, compared with 3 of 38 who did not (80% versus 8%, P 0.0001). Thyroid dysfunction occurred early (median, 42 days) in the pembrolizumab course, and a majority (6 of 10 patients) experienced brief, transient hyperthyroidism preceding the onset of hypothyroidism; no persistent hyperthyroidism occurred. Both hyperthyroidism and hypothyroidism were largely asymptomatic. Overall survival with pembrolizumab was significantly longer in subjects who developed thyroid dysfunction (hazard ratio, 0.29; 95% CI 0.09-0.94; P = 0.04).Thyroid dysfunction during pembrolizumab treatment of NSCLC is common and is characterized by early-onset, frequently preceded by transient hyperthyroidism, closely associated with anti-thyroid antibodies, and may be associated with improved outcomes. The presence of antibody-mediated toxicity in T-cell-directed therapy suggests an under-recognized impact of PD-1 biology in modulating humoral immunity.
- Published
- 2017
16. Immunophenotype and Response to Immunotherapy of RET-Rearranged Lung Cancers
- Author
-
Dazhi Liu, Joshua K. Sabari, Bob T. Li, Charles M. Rudin, Robin Guo, Natasha Rekhtman, Gregory J. Riely, Maria E. Arcila, Stephanie L. Wu, Marc Ladanyi, Ai Ni, Ryma Benayed, Terry Pak, Mark G. Kris, Joseph Montecalvo, Alexander Drilon, Michael Offin, Josiah D. Land, Larry W Buie, Darragh Halpenny, and Matthew D. Hellmann
- Subjects
Oncology ,0303 health sciences ,Cancer Research ,medicine.medical_specialty ,Chemotherapy ,Lung ,business.industry ,medicine.medical_treatment ,Immunotherapy ,Disease ,Immune checkpoint ,Article ,3. Good health ,Blockade ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Immunophenotyping ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,business ,030304 developmental biology - Abstract
RET rearrangements are identified in 1% to 2% of non-small-cell lung cancers (NSCLCs).1,2 In patients with advanced, RET-rearranged lung cancers, systemic therapy can be highly active. We demonstrated previously that pemetrexed-based chemotherapy can achieve an objective response rate of 45% and a median progression-free survival (PFS) of 19 months.3 Furthermore, the activity of targeted therapy has improved dramatically with the introduction of selective RET inhibitors to the clinic. In early-phase testing, objective response rates with LOXO-2924 and BLU-6675 are 68% (26 of 38) and 50% (seven of 14), respectively. These outcomes exceed the modest activity observed previously with multikinase inhibitors such as cabozantinib6 and vandetanib.7 In contrast, the activity of immunotherapy in RET-rearranged lung cancers has not been well characterized. This represents a clear unmet need, given that all prior regulatory approvals of immune checkpoint inhibitors, either alone or in combination with chemotherapy, and in stage III or IV disease, have technically included patients with RET-rearranged lung cancers.8,9 Furthermore, although increasing levels of programmed death-ligand 1 (PD-L1) expression and high tumor mutational burden (TMB) have been associated with benefit from immune checkpoint blockade,10 the immunophenotype of RET-rearranged lung cancers and the role of PD-L1 and TMB status in relation to benefit with immunotherapy remain poorly described. We set out to characterize these factors.
- Published
- 2019
17. Frequency and outcomes of brain metastases in patients with HER2-mutant lung cancers
- Author
-
Mackenzie L. Myers, Elena Pentsova, Maria E. Arcila, Mark G. Kris, Robert J. Young, Adrienne Boire, Ai Ni, Kathryn Beal, David R. Jones, Lisa M. DeAngelis, W. Victoria Lai, James M. Isbell, Helena A. Yu, Viviane Tabar, Gregory J. Riely, Mariza Daras, Charles M. Rudin, Bob T. Li, Emmet Jordan, Alexander Drilon, Daniel Feldman, David B. Solit, and M. Offin
- Subjects
Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Receptor, ErbB-2 ,medicine.disease_cause ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Internal medicine ,medicine ,Biomarkers, Tumor ,Odds Ratio ,Humans ,030212 general & internal medicine ,Epidermal growth factor receptor ,skin and connective tissue diseases ,Lung cancer ,neoplasms ,Aged ,Proportional Hazards Models ,Aged, 80 and over ,Mutation ,Lung ,biology ,Radiotherapy ,business.industry ,Brain Neoplasms ,Incidence ,Hazard ratio ,Odds ratio ,Oncogenes ,Middle Aged ,medicine.disease ,Prognosis ,Patient Outcome Assessment ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,biology.protein ,Female ,KRAS ,business ,Brain metastasis - Abstract
BACKGROUND Mutations in human epidermal growth factor receptor 2 (HER2; also known as ERBB2) are found in approximately 2% of lung adenocarcinomas. The frequency and clinical course of brain metastases in this oncogenic subset are ill defined. METHODS Baseline and subsequent development of brain metastases was evaluated in consecutive patients with HER2-mutant (n = 98), epidermal growth factor receptor (EGFR)-mutant (n = 200), and KRAS-mutant lung cancers (n = 200). RESULTS At metastatic diagnosis, the odds ratio (ORs) for brain metastases was similar for patients whose tumors harbored HER2 mutations (19%) in comparison with patients with KRAS mutations (24%; OR for HER2 vs KRAS, 0.7; P = .33) but lower compared to patients with EGFR mutations (31%; OR for HER2 vs EGFR, 0.5; P = .03). Patients with lung cancer and HER2 mutations developed more brain metastases on treatment than patients with KRAS mutations (28% vs 8%; hazard ratio [HR], 5.2; P
- Published
- 2019
18. Prognostic risk score for patients with relapsed or refractory chronic lymphocytic leukaemia treated with targeted therapies or chemoimmunotherapy: a retrospective, pooled cohort study with external validations
- Author
-
Asher Chanan-Khan, Jeffrey A. Jones, Danelle F James, Ai Ni, Guan Xing, Neil E. Kay, Mehrdad Mobasher, Anil Londhe, John C. Byrd, Richard R. Furman, John F. Seymour, Yeung-Chul Mun, Pankaj Bhargava, Jacob D. Soumerai, Lucille Ferrante, Mohamed Darif, Vijay Reddy, Angela Howes, Tait D. Shanafelt, Peter Hillmen, Thomas Stark, Jeffrey P. Sharman, Kari G. Rabe, Lyndah Dreiling, and Andrew D. Zelenetz
- Subjects
Oncology ,Male ,medicine.medical_specialty ,Databases, Factual ,Antineoplastic Agents ,Ofatumumab ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Hemoglobins ,0302 clinical medicine ,Piperidines ,Chemoimmunotherapy ,Risk Factors ,Internal medicine ,medicine ,Humans ,Survival rate ,Proportional Hazards Models ,Quinazolinones ,Randomized Controlled Trials as Topic ,Retrospective Studies ,Sulfonamides ,L-Lactate Dehydrogenase ,Venetoclax ,business.industry ,Adenine ,Retrospective cohort study ,Hematology ,Middle Aged ,Bridged Bicyclo Compounds, Heterocyclic ,Prognosis ,Leukemia, Lymphocytic, Chronic, B-Cell ,Survival Rate ,Pyrimidines ,chemistry ,Purines ,030220 oncology & carcinogenesis ,Ibrutinib ,Pyrazoles ,Female ,Immunotherapy ,Idelalisib ,business ,beta 2-Microglobulin ,030215 immunology ,Cohort study - Abstract
Summary Background Clinically validated prognostic models for overall survival do not exist for patients with relapsed or refractory chronic lymphocytic leukaemia (CLL) who are on targeted therapies. We aimed to create a prognostic model to identify high-risk individuals who do not achieve a good outcome with available targeted therapies. Methods In this retrospective, pooled cohort study, 2475 patients with CLL treated between June 22, 2012, and Sept 23, 2015, in six randomised trials of ibrutinib, idelalisib, and venetoclax, or at the Mayo Clinic CLL Database (MCCD) were included. Eligible patients had CLL, were previously treated, were aged 18 years or older, had ECOG performance status 0–1, and required further treatment as per the international workshop on CLL 2008 criteria. There was heterogeneity in other eligibility criteria. We evaluated 28 candidate factors known to affect the overall survival of these patients and applied univariate and multivariate analyses to derive the risk score in a training dataset (n=727) of patients treated with ibrutinib or chemoimmunotherapy. We validated the score in an internal-validation dataset (n=242) of patients treated with ibrutinib or chemoimmunotherapy and three external-validation datasets (idelalisib or chemoimmunotherapy dataset, n=897; venetoclax or chemoimmunotherapy dataset, n=389; and the MCCD [including patients treated with heterogeneous therapies], n=220), applying C-statistics as a measure of discrimination. Findings The derived model consisted of four factors (one point each; serum β2-microglobulin ≥5 mg/dL, lactate dehydrogenase >upper limit of normal, haemoglobin Interpretation We present the first validated risk score to predict overall survival in patients with relapsed or refractory CLL treated with targeted therapy. The model is applicable to patients treated with all currently approved targeted therapies (ibrutinib, idelalisib, and venetoclax) and chemoimmunotherapy. This tool allows the identification of a well defined cohort of previously treated patients with CLL who are at high risk of death, and could be used in future prospective trials to test therapeutic options for these patients with an unmet clinical need. Funding Lymphoma Research Foundation, Lymphoma Research Fund (Andrew D Zelenetz), and National Institutes of Health/National Cancer Institute.
- Published
- 2019
19. Exceptional responders with invasive mucinous adenocarcinomas: a phase 2 trial of bortezomib in patients with KRAS G12D-mutant lung cancers
- Author
-
Ai Ni, Maureen Kennedy, Anne Haughney-Siller, Linda Ahn, Gregory J. Riely, Elizabeth Panora, Adam J. Schoenfeld, Natasha Rekhtman, Anna Maria Litvak, Marc Ladanyi, Mark G. Kris, Helena A. Yu, Michelle S. Ginsberg, Maria E. Arcila, Kathryn C. Arbour, Vincent A. Miller, Alexander Drilon, and Joseph Montecalvo
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Lung Neoplasms ,Injections, Subcutaneous ,Antineoplastic Agents ,medicine.disease_cause ,Bortezomib ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Humans ,Prospective Studies ,Stage (cooking) ,Pack-year ,Adverse effect ,Lung cancer ,neoplasms ,030304 developmental biology ,Aged ,Aged, 80 and over ,0303 health sciences ,business.industry ,General Medicine ,Middle Aged ,medicine.disease ,lung adenocarcinoma ,Adenocarcinoma, Mucinous ,3. Good health ,respiratory tract diseases ,Clinical trial ,Treatment Outcome ,030220 oncology & carcinogenesis ,Mutation ,Proteasome inhibitor ,Female ,KRAS ,business ,medicine.drug ,Research Article - Abstract
KRAS G12D-mutant/p53-deficient non-small-cell lung cancer (NSCLC) models are dependent on the NF-κB pathway that can be down-regulated by the proteasome inhibitor bortezomib. Two exceptional responders were observed on prior clinical trials of bortezomib, both of whom had KRAS G12D-mutant NSCLC, prompting the initiation of this single-center phase 2 trial. Patients with advanced KRAS G12D-mutant NSCLC were eligible. Bortezomib was administered at 1.3 mg/m2 subcutaneously (days 1, 4, 8, 11; 21-d cycle) until progression or unacceptable toxicity. The primary objective was best objective response (RECIST v1.1). Sixteen patients with KRAS G12D-mutant lung adenocarcinomas were treated. Patients had a median pack year smoking history of 4 (range 0–45). A partial response (PR) was observed in one patient (−66% from baseline) and stable disease in five patients on the first stage of this study (overall response rate of 6%, 95% CI: 0.2–30.2), and further patients were not accrued. The median progression-free survival was 1 mo (95% CI: 1–6). The median overall survival was 13 mo (95% CI: 6–NA). The most common treatment-related adverse events were fatigue (38%) and diarrhea (26%). TP53 status did not predict response on exploratory testing. Of note, the patient with a PR had a unique subtype of lung adenocarcinoma—invasive mucinous adenocarcinomas (IMA)—and had rapid clinical improvement and substantial disease regression, which was also previously observed in two other patients with advanced KRAS G12D-mutant lung cancer with IMAs who received bortezomib on separate clinical trials. Exceptional responses to bortezomib can be achieved in KRAS G12D-mutant NSCLCs. KRAS G12D mutation alone, however, is not a robust predictor of response. Further evaluation should only be performed after further elucidation of other factors such as co-occurring alterations and histologic subtype such as IMA that may predict sensitivity to therapy.
- Published
- 2019
20. A phase II study of axitinib (AG-013736) in patients with incurable adenoid cystic carcinoma
- Author
-
Cristina R. Antonescu, Camelia Sima, Luc G. T. Morris, David G. Pfister, Ai Ni, Alan Loh Ho, Nora Katabi, Snjezana Dogan, Grace Cullen, Matthew G. Fury, Raghu Chandramohan, Eric J. Sherman, Shrujal S. Baxi, Lara Dunn, and Sofia Haque
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Indazoles ,Axitinib ,Drug-Related Side Effects and Adverse Reactions ,Oncogene Proteins, Fusion ,Adenoid cystic carcinoma ,medicine.drug_class ,Phases of clinical research ,Context (language use) ,Disease-Free Survival ,Tyrosine-kinase inhibitor ,Proto-Oncogene Proteins c-myb ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,MYB ,Protein Kinase Inhibitors ,Aged ,Dose-Response Relationship, Drug ,business.industry ,Imidazoles ,High-Throughput Nucleotide Sequencing ,Original Articles ,Hematology ,Middle Aged ,medicine.disease ,Carcinoma, Adenoid Cystic ,NFI Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Chromosomes, Human, Pair 4 ,business ,Progressive disease ,medicine.drug - Abstract
Background Recurrent/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. Patients and methods This 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. Results Thirty-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). Conclusions Although 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.
- Published
- 2016
21. Prognostic value of baseline metabolic tumor volume measured on (18)F-fluorodeoxyglucose positron emission tomography/computed tomography in melanoma patients treated with ipilimumab therapy
- Author
-
Heiko Schöder, Kimiteru Ito, Ai Ni, Wolfgang A. Weber, Rebecca Teng, John L. Humm, and Jedd D. Wolchok
- Subjects
Oncology ,Adult ,Male ,medicine.medical_specialty ,Multivariate analysis ,medicine.medical_treatment ,Ipilimumab ,Article ,030218 nuclear medicine & medical imaging ,Fluorodeoxyglucose positron emission tomography ,03 medical and health sciences ,0302 clinical medicine ,Fluorodeoxyglucose F18 ,Internal medicine ,Positron Emission Tomography Computed Tomography ,medicine ,Humans ,Radiology, Nuclear Medicine and imaging ,Melanoma ,Aged ,Retrospective Studies ,Chemotherapy ,business.industry ,General Medicine ,Metabolic tumor volume ,Immunotherapy ,Middle Aged ,medicine.disease ,Prognosis ,Survival Analysis ,Tumor Burden ,CTLA-4 ,030220 oncology & carcinogenesis ,Female ,business ,medicine.drug - Abstract
Ipilimumab induces durable remission in about 15–20% of patients with metastatic melanoma. However, reliable predictors of response to ipilimumab are currently lacking. Whole-body metabolic tumor volume (wMTV) has been shown to be a strong prognostic factor in a variety of malignancies treated with chemotherapy, but few results have been reported for patients treated with immunotherapy. The purpose of this study was to investigate the prognostic value of wMTV and other metabolic parameters from baseline 18F-FDG PET/CT scans in patients with melanoma being treated with ipilimumab. The prognostic impact of wMTV, as well as mean standardized uptake values and total lesion glycolysis, was evaluated in 142 consecutive patients with melanoma treated with single-agent ipilimumab therapy. Metabolic parameters were dichotomized by their respective medians and correlated with overall survival (OS). In addition, the prognostic value of metabolic parameters combined with known clinical prognostic factors was evaluated in multivariate analyses. The median OS time in all patients was 14.7 months (95% CI 10.45–18.93 months). wMTV was a strong independent prognostic factor for OS (p = 0.001). The median survival in patients with a metabolic volume above the median was 10.8 months (95% CI 5.88–15.81 months) as compared with 26.0 months (95% CI 3.02–49.15 months) in patients with an MTV below the median. A multivariate model including wMTV and known clinical prognostic factors, such as age and the presence of brain metastases, further improved the identification of patient subgroups with different OS times. wMTV appears to be a strong independent prognostic factor in melanoma patients treated with ipilimumab, and can be determined semiautomatically from routine 18F- FDG PET/CT scans. wMTV, combined with clinical prognostic factors, could be used to personalize immunotherapy and in future clinical studies.
- Published
- 2018
22. Afatinib in patients with metastatic or recurrent HER2-mutant lung cancers: a retrospective international multicentre study
- Author
-
Maria E. Arcila, Andrew J. Plodkowski, John Kavanagh, Nick Pavlakis, Joshua K. Sabari, Roberto Ferrara, Stephen Clarke, Tristan A. Barnes, Julien Mazieres, Charles M. Rudin, Frances A. Shepherd, Oliver Gautschi, Mark G. Kris, Solange Peters, Louisiane Lebas, Bob T. Li, Alexander Drilon, Natasha B. Leighl, Ai Ni, W. Victoria Lai, and Julie Milia
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Receptor, ErbB-2 ,Afatinib ,Adenocarcinoma of Lung ,Antineoplastic Agents ,Article ,03 medical and health sciences ,0302 clinical medicine ,Stable Disease ,Trastuzumab ,Internal medicine ,medicine ,Humans ,In patient ,skin and connective tissue diseases ,Aged ,Retrospective Studies ,Aged, 80 and over ,Lung ,business.industry ,International Agencies ,Middle Aged ,medicine.disease ,Prognosis ,Confidence interval ,Survival Rate ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Female ,Pertuzumab ,Neoplasm Recurrence, Local ,business ,Progressive disease ,medicine.drug ,Follow-Up Studies - Abstract
Introduction HER2 mutations occur in 1–3% of lung adenocarcinomas. With increasing use of next-generation sequencing at diagnosis, more patients with HER2-mutant tumours present for treatment. Few data are available to describe the clinical course and outcomes of these patients when treated with afatinib, a pan-HER inhibitor. Methods We identified patients with metastatic or recurrent HER2-mutant lung adenocarcinomas treated with afatinib among seven institutions across Europe, Australia, and North America between 2009 and 2017. We determined the partial response rate to afatinib, types of HER2 mutations, duration of response, time on treatment, and survival. Results We collected information on 27 patients with stage IV or recurrent HER2-mutant lung adenocarcinomas treated with afatinib. Of 23 patients evaluable for response, three partial responses were noted (13%, 95% confidence interval [CI] 4–33%). In addition, 57% of patients (13/23) had stable disease, and 30% (7/23) had progressive disease. We documented partial responses in patients with HER2 exon 20 insertions, including two with YVMA insertion and one with VAG insertion. Two patients with partial responses were previously treated with trastuzumab and pertuzumab. Median duration of response to afatinib was 6 months (range 5–10); median time on treatment was 3 months (range 1–30) and median overall survival from the date of diagnosis of metastatic or recurrent disease was 23 months (95% CI 18–53 months). Conclusions Afatinib is modestly active in patients with HER2-mutant lung adenocarcinomas, including responses after progression on prior HER2-targeted therapies. However, investigations into the biology of HER2-mutant lung adenocarcinomas and development of better HER2-directed therapies are warranted.
- Published
- 2018
23. Ado-Trastuzumab Emtansine for Patients With HER2-Mutant Lung Cancers: Results From a Phase II Basket Trial
- Author
-
José Baselga, Alexander Drilon, Gregory J. Riely, Sarit Schwartz, Maurizio Scaltriti, Zachary T. Olah, Edyta B. Brzostowski, Ronglai Shen, Fabiola Cecchi, Stephen Clarke, Michelle S. Ginsberg, David M. Hyman, Helen Won, Maria E. Arcila, Ai Ni, Gary A. Ulaner, Daniel Feldman, David B. Solit, Nick Pavlakis, Mark G. Kris, Todd Hembrough, Michael Offin, Charles M. Rudin, Bob T. Li, Darren J. Buonocore, and Michael F. Berger
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Pathology ,Lung Neoplasms ,Receptor, ErbB-2 ,Adenocarcinoma of Lung ,Ado-Trastuzumab Emtansine ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Internal medicine ,medicine ,Clinical endpoint ,Humans ,Maytansine ,skin and connective tissue diseases ,Aged ,Lung ,Errata ,medicine.diagnostic_test ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,Trastuzumab ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Response Evaluation Criteria in Solid Tumors ,030220 oncology & carcinogenesis ,Mutation ,Toxicity ,Cohort ,Immunohistochemistry ,Female ,business ,Fluorescence in situ hybridization - Abstract
Purpose Human epidermal growth factor receptor 2 ( HER2, ERBB2)–activating mutations occur in 2% of lung cancers. We assessed the activity of ado-trastuzumab emtansine, a HER2-targeted antibody-drug conjugate, in a cohort of patients with HER2-mutant lung cancers as part of a phase II basket trial. Patients and Methods Patients received ado-trastuzumab emtansine at 3.6 mg/kg intravenously every 3 weeks until progression. The primary end point was overall response rate using Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. A Simon two-stage optimal design was used. Other end points included progression-free survival and toxicity. HER2 testing was performed on tumor tissue by next generation sequencing, fluorescence in situ hybridization, immunohistochemistry, and protein mass spectrometry. Results We treated 18 patients with advanced HER2-mutant lung adenocarcinomas. The median number of prior systemic therapies was two (range, zero to four prior therapies). The partial response rate was 44% (95% CI, 22% to 69%), meeting the primary end point. Responses were seen in patients with HER2 exon 20 insertions and point mutations in the kinase, transmembrane, and extracellular domains. Concurrent HER2 amplification was observed in two patients. HER2 immunohistochemistry ranged from 0 to 2+ and did not predict response, and responders had low HER2 protein expression measured by mass spectrometry. The median progression-free survival was 5 months (95% CI, 3 to 9 months). Toxicities included grade 1 or 2 infusion reactions, thrombocytopenia, and elevated hepatic transaminases. No patient stopped therapy as a result of toxicity or died on study. Conclusion Ado-trastuzumab emtansine is an active agent in patients with HER2-mutant lung cancers. This is the first positive trial in this molecular subset of lung cancers. Further use and study of this agent are warranted.
- Published
- 2018
24. Vemurafenib Redifferentiation of BRAF Mutant, RAI-Refractory Thyroid Cancers
- Author
-
Stephanie Fish, Eric J. Sherman, Jamie Walters, R. Michael Tuttle, Steven M. Larson, Ronald Ghossein, Vatche Tchekmedyian, David G. Pfister, Ravinder K. Grewal, Lara Dunn, James A. Fagin, Ai Ni, Keith S. Pentlow, Laura Boucai, Alan L. Ho, Venkatraman E. Seshan, Sofia Haque, Duan Li, Shrujal S. Baxi, Jeffrey A. Knauf, and Mona M. Sabra
- Subjects
0301 basic medicine ,Oncology ,MAPK/ERK pathway ,Adult ,Male ,Proto-Oncogene Proteins B-raf ,medicine.medical_specialty ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Clinical Biochemistry ,Context (language use) ,Pilot Projects ,Biochemistry ,Radiation Tolerance ,Iodine Radioisotopes ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Refractory ,Internal medicine ,Positron Emission Tomography Computed Tomography ,medicine ,Humans ,Thyroid Neoplasms ,Vemurafenib ,Thyroid cancer ,Protein Kinase Inhibitors ,Thyrotropin Alfa ,Aged ,business.industry ,Biochemistry (medical) ,Thyroid ,Cancer ,Cell Differentiation ,Cell Dedifferentiation ,Middle Aged ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Thyroid Cancer, Papillary ,030220 oncology & carcinogenesis ,Thyroglobulin ,Female ,business ,medicine.drug - Abstract
Context BRAFV600E mutant thyroid cancers are often refractory to radioiodine (RAI). Objectives To investigate the utility and molecular underpinnings of enhancing lesional iodide uptake with the BRAF inhibitor vemurafenib in patients with RAI-refractory (RAIR). Design This was a pilot trial that enrolled from June 2014 to January 2016. Setting Academic cancer center. Patients Patients with RAIR, BRAF mutant thyroid cancer. Intervention Patients underwent thyrotropin-stimulated iodine-124 (124I) positron emission tomography scans before and after ~4 weeks of vemurafenib. Those with increased RAI concentration exceeding a predefined lesional dosimetry threshold (124I responders) were treated with iodine-131 (131I). Response was evaluated with imaging and serum thyroglobulin. Three patients underwent research biopsies to evaluate the impact of vemurafenib on mitogen-activated protein kinase (MAPK) signaling and thyroid differentiation. Main outcome measure The proportion of patients in whom vemurafenib increased RAI incorporation to warrant 131I. Results Twelve BRAF mutant patients were enrolled; 10 were evaluable. Four patients were 124I responders on vemurafenib and treated with 131I, resulting in tumor regressions at 6 months. Analysis of research tumor biopsies demonstrated that vemurafenib inhibition of the MAPK pathway was associated with increased thyroid gene expression and RAI uptake. The mean pretreatment serum thyroglobulin value was higher among 124I responders than among nonresponders (30.6 vs 1.0 ng/mL; P = 0.0048). Conclusions Vemurafenib restores RAI uptake and efficacy in a subset of BRAF mutant RAIR patients, probably by upregulating thyroid-specific gene expression via MAPK pathway inhibition. Higher baseline thyroglobulin values among responders suggest that tumor differentiation status may be a predictor of vemurafenib benefit.
- Published
- 2018
25. Expression of PD-L1 and other immunotherapeutic targets in thymic epithelial tumors
- Author
-
Andreas Rimner, Jarushka Naidoo, Andre L. Moreira, Ai Ni, Paul B. Robbins, Natasha Rekhtman, Keith Steele, Gregory J. Riely, Joyson Joseph Karakunnel, Kathryn C. Arbour, Matthew D. Hellmann, and James Huang
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Cancer Treatment ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Lung and Intrathoracic Tumors ,B7-H1 Antigen ,White Blood Cells ,0302 clinical medicine ,Cancer immunotherapy ,Thymic Tumors ,Animal Cells ,Medicine and Health Sciences ,Tumor Microenvironment ,CTLA-4 Antigen ,Neoplasms, Glandular and Epithelial ,lcsh:Science ,Hepatitis A Virus Cellular Receptor 2 ,Thymic carcinoma ,Staining ,Multidisciplinary ,biology ,T Cells ,CD137 ,Cell Staining ,3. Good health ,Membrane Staining ,Oncology ,030220 oncology & carcinogenesis ,Female ,Immunotherapy ,Cellular Types ,Research Article ,Thymoma ,Immune Cells ,Immunology ,Cytotoxic T cells ,Research and Analysis Methods ,Carcinomas ,Cancer Immunotherapy ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,PD-L1 ,Glucocorticoid-Induced TNFR-Related Protein ,medicine ,Biomarkers, Tumor ,Humans ,Molecular Biology Techniques ,Molecular Biology ,Tumor microenvironment ,Blood Cells ,business.industry ,Tumor-infiltrating lymphocytes ,lcsh:R ,Cancers and Neoplasms ,Biology and Life Sciences ,Cell Biology ,Thymus Neoplasms ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Survival Analysis ,Lymphocyte Subsets ,030104 developmental biology ,Specimen Preparation and Treatment ,Tissue Array Analysis ,biology.protein ,lcsh:Q ,Clinical Immunology ,Clinical Medicine ,business ,Cloning - Abstract
Introduction The thymus is a critical organ for the development of the adaptive immune system and thymic epithelial tumors (TETs; thymomas and thymic carcinomas) are often associated with auto-immune paraneoplastic conditions. However, the immunobiology of TETs is not well described. An evaluation of the tumor microenvironment, with particular focus on expression of immunotherapeutic targets, may facilitate and prioritize development of immunotherapy strategies for patients with TETs. Methods Tumor tissues from 23 patients with WHO Type B2/B3 thymoma (n = 12) and thymic carcinoma (n = 11) were identified and clinical outcomes were annotated. The expression of membranous PD-L1 on tumor cells, CD3+ and CD8+ tumor infiltrating lymphocytes (TILs), co-stimulatory (CD137, GITR, ICOS), and co-inhibitory immune checkpoint molecules (PD-1, CTLA-4, TIM-3) were assessed semi-quantitatively using immunohistochemistry. Results PD-L1 positivity (≥ 25% of tumor membrane expression) was frequent in TETs (15/23, 65%), more common in thymomas compared to thymic carcinomas (p
- Published
- 2017
26. Prognostic impact of TTF-1 expression in patients with stage IV lung adenocarcinomas
- Author
-
Matthew D. Hellmann, Ai Ni, Sutirtha Datta, William D. Travis, Linda Ahn, Juliana Schilsky, N. Rekhtman, Mark G. Kris, and Jamie E. Chaft
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,Lung Neoplasms ,medicine.medical_treatment ,Thyroid Nuclear Factor 1 ,Gene Expression ,Targeted therapy ,0302 clinical medicine ,Aged, 80 and over ,biology ,Combination chemotherapy ,respiratory system ,Middle Aged ,Prognosis ,Immunohistochemistry ,Survival Rate ,Pemetrexed ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Antibody ,medicine.drug ,Pulmonary and Respiratory Medicine ,Adult ,medicine.medical_specialty ,endocrine system ,Adolescent ,Adenocarcinoma of Lung ,Adenocarcinoma ,Article ,03 medical and health sciences ,Young Adult ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Lung cancer ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,Chemotherapy ,Lung ,business.industry ,medicine.disease ,030104 developmental biology ,Mutation ,biology.protein ,business - Abstract
OBJECTIVES: Thyroid transcription factor 1 (TTF-1) is routinely tested in the diagnostic evaluation of suspected lung cancers, is commonly expressed by lung adenocarcinomas, and may modulate lung cancer biology. We examined the role of TTF-1 as a predictive and prognostic marker in patients with advanced lung adenocarcinomas. MATERIALS AND METHODS: We analyzed clinical, pathologic, and molecular features, treatments received, and overall survival obtained from the medical records of 479 consecutive patients at a single site with stage IV lung adenocarcinomas and evaluable TTF-1 expression. TTF-1 expression was determined by immunohistochemistry using antibody 8G7G3/1. RESULTS AND CONCLUSION: TTF-1 expression was evaluable in 479 (75%) of all patients reviewed, and was positive in 383 (80%, 95% CI 76 to 83%). Clinicopathologic features were similar between TTF-1 positive and TTF-1 negative tumors, except EGFR mutations were more common in TTF-1 positive cases (24% vs 6%, p/= 80% (HR 0.62, p=0.0003) and receipt of first-line combination chemotherapy or targeted therapy (HR relative to single agent chemotherapy 0.59, p=0.05 and 0.51, p=0.05 respectively). Both patients with TTF-1 positive and TTF-1 negative cancers had longer durations of initial therapy when treated with pemetrexed-based chemotherapy. In patients with advanced lung adenocarcinomas, TTF-1 expression is associated with better survival but is not predictive of distinct benefit from pemetrexed-based chemotherapy.
- Published
- 2017
27. REAL-WORLD ASSESSMENT OF PRACTICE EFFICIENCY WITH THE INTRODUCTION OF SUBCUTANEOUS RITUXIMAB
- Author
-
Arliene Ravelo, Tu My To, Keith L Dawson, Sheila Shapouri, J. Schade, Ai Ni, Annie Qiu, Matthew J. Matasar, and Esther Drill
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Follicular lymphoma ,Hematology ,General Medicine ,medicine.disease ,Internal medicine ,Medicine ,Rituximab ,business ,Intensive care medicine ,medicine.drug - Abstract
e18025 Background: Rituximab (R), available as an intravenous (IV; R-IV) infusion or subcutaneous (SC; R-SC) injection, is used in the treatment of follicular lymphoma (FL), diffuse large B-cell lymphoma (DLBCL), and chronic lymphocytic leukemia (CLL). This study evaluated real-world practice efficiency changes associated with the adoption of R-SC by studying differences in chair time by route of administration. Methods: We conducted a retrospective analysis of practice care delivery measures before and after adoption of R-SC at Memorial Sloan Kettering Cancer Center (MSKCC). Data for patients (pts) with FL, DLBCL, or CLL receiving R-based therapy from September 2016 to September 2018 were extracted from the electronic medical record. A linear mixed effect multivariate model with random intercept was used to analyze the association between treatment type (R-IV vs R-SC) and chair time (defined as the difference in pt room-in and room-out times) in the year prior to and following R-SC adoption at MSKCC. Model covariates included treatment time and location, therapy type (monotherapy vs combination), and pt demographics. Given the prolonged infusion time, patients’ first dose of R-IV was excluded from the analysis. Results: Data were collected during 6744 visits (3018 visits prior to R-SC adoption and 3726 after) for 1503 pts receiving R. Pts receiving R-IV combination therapy had a mean chair time of 203 minutes (min); overall, R-SC injection reduced chair time by a mean of 92 min (p < 0.001 vs R-IV). Monotherapy, regardless of route, reduced chair time by a mean of 30 min (p < 0.001) compared with combination therapy, and mean chair time was further reduced by 39 min (p < 0.001) for R-SC pts receiving monotherapy. Reductions in chair time increased over time following initial adoption of R-SC (p = 0.042), and were greater at the lymphoma-specific site than multispecialty oncology infusion centers (p < 0.001). Conclusions: Adoption of R-SC results in substantial time savings for both the pt and health system as measured by reduced chair time and improved pt throughput. Given increasing constraints on infusion chair space, increased utilization of R-SC may improve practice efficiency and pt access to care.
- Published
- 2019
28. Risk of breast implant-associated anaplastic large cell lymphoma (BIA-ALCL) in a cohort of 3,546 women prospectively followed after receiving textured breast implants
- Author
-
Peter G. Cordeiro, Ahmet Dogan, Ai Ni, Qun-Ying Hu, Natasha Galasso, Nivetha Ganesan, Steven M. Horwitz, and Paola Ghione
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Capsule ,medicine.disease ,Lymphoma ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,hemic and lymphatic diseases ,030220 oncology & carcinogenesis ,Internal medicine ,Cohort ,Breast implant ,medicine ,business ,Anaplastic large-cell lymphoma ,030215 immunology - Abstract
1565 Background: Breast implant-associated anaplastic large-cell lymphoma (BIA-ALCL) is a rare subtype of T-cell lymphoma, developing in in the fluid or capsule surrounding breast implants, primarily or exclusively in those with textured surfaces. Several prior series have estimated the risk of BIA- ALCL at 1/6920 - 1/3800 women in retrospectively defined cohorts (from diagnosed cases within national or pathology databases), approximating the population at risk from sales records or other estimates (Sirinvasa 2017; Loch-Wilkinson 2017; de Boer 2018). Methods: A prospective cohort study was conducted in the population that underwent breast reconstruction by a single surgeon at Memorial Sloan Kettering Cancer Center (MSKCC) from April 1993 to December 2017. Patients had long-term follow-up, and events related to implants were prospectively recorded. We identified all cases of BIA-ALCL by cross-checking data from internal clinical records, pathology records, and outside reports. Incidence rate per person-years and cumulative incidence when accounting for competing risk were calculated. 134 women who received smooth-surface implants were excluded from the analysis, since these implants have not been associated with BIA-ALCL. Results: From 1993 to 2017, 3546 patients underwent 6023 breast reconstructions using textured surface implants. All reconstructions were performed by a single surgeon (PGC) on patients enrolled in this study. To identify BIA-ALCL occurrence, clinical and pathological data were assessed from a prospective database. Median follow-up was 7 years (range, 3 days - 24.7 years). Eight women developed ALCL after a median exposure of 11.2 years (range, 8.3-15.8 years). Overall risk of BIA-ALCL in this cohort was 0.294 cases per 1000 person-years (1/443 women). Conclusions: This study, evaluating the risk of women with textured breast implants from a prospective database with long-term follow-up, demonstrated that the incidence rate of BIA-ALCL may be higher than previously reported. These results can help inform implant choice for women undergoing breast reconstruction.
- Published
- 2019
29. A phase 1/2 study of osimertinib and bevacizumab as initial treatment for patients with metastatic EGFR-mutant lung cancers
- Author
-
Rachel Kim, Alex Makhnin, Robert J. Young, Sara A. Hayes, Ai Ni, Helena Alexandra Yu, Gregory J. Riely, Mark G. Kris, and Linda Su Hyun Ahn
- Subjects
Cancer Research ,Lung ,Bevacizumab ,business.industry ,Mutant ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Initial treatment ,Osimertinib ,Erlotinib ,business ,030215 immunology ,medicine.drug - Abstract
9086 Background: Osimertinib (osi) demonstrated improved progression-free survival (PFS) over erlotinib as initial treatment (trmt) for EGFR-mutant (EGFR+) lung cancers. The addition of bevacizumab (bev) to erlotinib as initial trmt resulted in improved PFS compared to erlotinib alone (16 vs 10 months, HR 0.41). The phase 1 study of osi and bev confirmed the ability to combine osi and bev at full doses. Methods: The phase 2 study is assessing safety and efficacy of the combination. The primary endpoint is PFS at 12 months; secondary endpoints include overall response (ORR), overall survival (OS), and CNS PFS. All pts had interval CNS as well as systemic imaging. Pre-treatment and post-progression (PD) tumor tissue and interval plasma samples are being collected to identify mechanisms of resistance (MOR) and for biomarker assessment. Results: From Nov 2016 to May 2018, 49 pts were enrolled, including 6 pts from the phase 1. Median age: 60; Women 69%; Never-smokers 65%. 13 pts had CNS metastases (9 untreated, 5 measurable) prior to study initiation. 49 pts are eligible for response; 34/49 pts had a confirmed partial response (PR)(ORR 69%). PFS at 12 months is 0.70 (95% CI: 0.57-0.84), with 8/49 pts on study without PD for less than 12 months. All pts with measurable CNS disease had a PR in the CNS; PD in the CNS was uncommon (17%). 24 pts remain on study without PD; 2 are being treated beyond PD. Reasons for study discontinuation include PD (n = 16), resection of all sites of disease (n = 3), withdrawal of consent (n = 3), unrelated death (n = 2), toxicity (n = 1). The most frequent trtmt-related adverse events (any grade) were thrombocytopenia (61%), diarrhea (57%), hypertension (55%), and rash (47%). 24% required a dose reduction of osi, 18% discontinued bev and median doses of bev was 17. 9 pts have paired pre-trtmt and post-progression tissue biopsies; MOR identified include squamous cell (n = 2) and small cell (n = 1) transformation, PTEN loss, and CCNE amplification. Conclusions: Combination osi and bev was well-tolerated and efficacy to date supports further evaluation. Results of secondary endpoints including PFS, mechanisms of resistance, cfDNA data are forthcoming. A randomized study of osi compared to osi and bev is planned (EA5182). Supported by AstraZeneca Clinical trial information: NCT02803203.
- Published
- 2019
30. Risk Model for Overall Survival for Patients with Relapsed/Refractory Chronic Lymphocytic Leukemia: Validated for Patients on Ibrutinib, Idelalisib, Venetoclax, or Chemoimmunotherapy
- Author
-
Yeung-Chul Mun, Mehrdad Mobasher, Kari G. Chaffee, Asher Chanan-Khan, Vijay Reddy, Neil E. Kay, Jeffrey A. Jones, Tait D. Shanafelt, Ai Ni, Danelle F. James, Peter Hillmen, John C. Byrd, Jacob D. Soumerai, Richard R. Furman, John F. Seymour, Mohamed Darif, Angela Howes, Guan Xing, Thomas Stark, Andrew D. Zelenetz, Pankaj Bhargava, Lyndah Dreiling, Lucille Ferrante, Anil Londhe, and Jeff P. Sharman
- Subjects
Oncology ,Bendamustine ,medicine.medical_specialty ,business.industry ,Venetoclax ,Chronic lymphocytic leukemia ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Ofatumumab ,Biochemistry ,chemistry.chemical_compound ,chemistry ,Chemoimmunotherapy ,Internal medicine ,Ibrutinib ,Medicine ,Rituximab ,business ,Idelalisib ,medicine.drug - Abstract
Introduction: Several prognostic models for overall survival (OS) in chronic lymphocytic leukemia (CLL) have been developed, but none were derived for patients (pts) with relapsed or refractory (R/R) CLL treated in the era of novel agents. We used a comprehensive approach to derive and validate a novel risk model for OS in 2,475 pts with R/R CLL who received therapy in randomized phase 3 trials evaluating ibrutinib (IBR), idelalisib (IDELA), and venetoclax (VEN) vs control arms, or the Mayo Clinic CLL Database (MCCD). Methods: In collaboration with Janssen, Gilead, Pharmacyclics, and Genentech/Roche, the analysis included 2,475 pts from 6 multicenter randomized phase 3 trials or the MCCD. Trials included were: IBR plus bendamustine-rituximab (BR) vs placebo plus BR (HELIOS: NCT01611090), IBR vs ofatumumab (RESONATE: NCT01578707), IDELA plus BR vs placebo plus BR (Study 115: NCT01569295); IDELA plus rituximab vs placebo plus rituximab (Study 116: NCT01539512); IDELA plus ofatumumab vs ofatumumab (Study 119: NCT01659021); VEN plus rituximab vs BR (MURANO: NCT02005471). All pts had R/R CLL and required treatment by iwCLL criteria. The model-building dataset (n=969) included pts from HELIOS and RESONATE, and was randomly assigned to the IBR/chemoimmunotherapy (CIT) training dataset (n=727) and IBR/CIT internal-validation dataset(n=242). Three independent external validation datasets included IDELA/CIT (n=897), VEN/CIT (n=389), and MCCD (n=220). We applied univariate and multivariate analyses (MVA) to 28 candidate clinical/laboratory and genetic prognostic factors to derive the risk model in the training dataset. The primary endpoint was OS. We assessed the need for separate models for targeted agents by interactions with treatment in univariate analyses. We proceeded to develop a single model for all pts with R/R CLL, as only one candidate factor (del(13q), interaction p value We evaluated cutoffs for dichotomized covariates and for definition of risk groups after the final factors were selected. We fit a Cox regression on the training and internal/external validation datasets using the risk categories as the covariate to test the difference among the groups, and calculated the C-statistic as a measure of discrimination. Results: Of 11 selected factors with a significant univariate effect on OS (p We excluded number of prior therapies from the model, reasoning that its optimal cutoff might change with advances in CLL therapies. IGHV did not remain significant (p=0.0592) in MVA of 5 remaining prognostic factors. Notably, del(17p) was not independently prognostic for OS. The final risk model consisted of 4 prognostic factors: B2M ≥5 mg/L, LDH elevated, HGB Our risk model was prognostic for OS in the IBR/CIT training dataset (C-statistic 0.74 [95% CI 0.60-0.85]; log-rank p Conclusions: We present the first validated risk model for OS in R/R CLL in the era of targeted therapies. Our model consists of 4 readily available factors (B2M ≥5 mg/L, LDH elevated, HGB Figure. Figure. Disclosures Darif: Jannsen: Employment. Londhe:Jannsen: Employment. Xing:Gilead Sciences, Inc.: Employment. Mun:Genentech: Employment, Equity Ownership. Kay:Cytomx Therapeutics: Membership on an entity's Board of Directors or advisory committees; Pharmacyclics: Membership on an entity's Board of Directors or advisory committees, Research Funding; Tolero Pharmaceuticals: Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Membership on an entity's Board of Directors or advisory committees; Infinity Pharm: Membership on an entity's Board of Directors or advisory committees; Agios Pharm: Membership on an entity's Board of Directors or advisory committees; Morpho-sys: Membership on an entity's Board of Directors or advisory committees; Acerta: Research Funding; Gilead: Membership on an entity's Board of Directors or advisory committees; Janssen: Membership on an entity's Board of Directors or advisory committees. Shanafelt:Mayo Clinic: Patents & Royalties: Physician Well-being Index, Medical Student Well-being Index, Well-being index; GlaxoSmithKline: Research Funding; Genentech: Research Funding; Pharmacyclics: Research Funding; Janssen: Research Funding; Abbvie: Research Funding; Celgene: Research Funding. Furman:Loxo Oncology: Consultancy; Pharmacyclics LLC, an AbbVie Company: Consultancy; Sunesis: Consultancy; TG Therapeutics: Consultancy; Verastem: Consultancy; Gilead: Consultancy; Incyte: Consultancy, Other: DSMB; Acerta: Consultancy, Research Funding; Genentech: Consultancy; Janssen: Consultancy; AbbVie: Consultancy. Hillmen:Janssen: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Abbvie: Honoraria, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Research Funding; Pharmacyclics: Research Funding; Alexion Pharmaceuticals, Inc: Consultancy, Honoraria; Novartis: Research Funding; Gilead Sciences, Inc.: Honoraria, Research Funding; F. Hoffmann-La Roche Ltd: Research Funding; Acerta: Membership on an entity's Board of Directors or advisory committees. Sharman:Acerta: Consultancy, Research Funding; Pharmacyclics, an AbbVie Company: Consultancy, Research Funding. Seymour:AbbVie: Consultancy, Honoraria, Research Funding; Celgene: Consultancy; F. Hoffmann-La Roche Ltd: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Genentech Inc: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Janssen: Honoraria, Research Funding. Jones:Celgene: Employment, Equity Ownership. Ferrante:Jannsen: Employment. Dreiling:Gilead: Employment. Mobasher:F. Hoffmann-La Roche Ltd: Other: Ownership interests non-PLC; Genentech Inc: Employment. Stark:Genentech: Employment. Reddy:Actinium Pharmaceuticals: Employment, Equity Ownership; Pharmacyclics: Employment. Howes:Janssen: Employment. James:Pharmacyclics: Employment. Bhargava:Gilead: Employment. Zelenetz:Novartis/Sandoz: Consultancy; Abbvie: Research Funding; Celgene: Consultancy; Amgen: Consultancy; Gilead: Consultancy, Research Funding; AstraZeneca: Consultancy; Genentech/Roche: Consultancy, Research Funding.
- Published
- 2018
31. Phase I/II clinical trial of ibrutinib and buparlisib in relapsed/refractory diffuse large B-cell lymphoma, mantle cell lymphoma, and follicular lymphoma
- Author
-
Joanna Dicostanzo, Heiko Schöder, Paul A. Hamlin, Devin Callan, Craig H. Moskowitz, Alison J. Moskowitz, John F. Gerecitano, David J. Straus, Ai Ni, Matthew J. Matasar, Stephanie De Frank, Anita Kumar, Andrew D. Zelenetz, Dana W.Y. Tsui, Anas Younes, Caitlin Stewart, Audrey Hamilton, Connie Lee Batlevi, Pamela Drullinsky, and Jürgen Rademaker
- Subjects
0301 basic medicine ,Cancer Research ,Buparlisib ,Follicular lymphoma ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,medicine ,Bruton's tyrosine kinase ,biology ,business.industry ,medicine.disease ,humanities ,Lymphoma ,030104 developmental biology ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,Ibrutinib ,biology.protein ,Cancer research ,Mantle cell lymphoma ,business ,Diffuse large B-cell lymphoma ,Tyrosine kinase - Abstract
7520Background: Bruton’s tyrosine kinase (BTK) and PI3K inhibitors synergize effectively in both in vitro and in vivo models of B-cell non-Hodgkin lymphoma (Griner, et al, PNAS, 2014; Erdmann et al...
- Published
- 2018
32. Prognostic relevance of tumor sequencing in metastatic lung adenocarcinomas
- Author
-
Charles M. Rudin, Ahmet Zehir, Venkatraman E. Seshan, Marc Ladanyi, Ai Ni, Gregory J. Riely, Maria E. Arcila, Axel Martin, Kathryn C. Arbour, Michael F. Berger, Matthew D. Hellmann, Mark G. Kris, Ryan Ptashkin, Arshi Arora, Emmet Jordan, Ronglai Shen, and David B. Solit
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,business.industry ,medicine.disease ,DNA sequencing ,Rapid identification ,medicine.anatomical_structure ,Internal medicine ,medicine ,Relevance (information retrieval) ,business ,Lung cancer - Abstract
9049Background: Routine next generation sequencing (NGS) testing for patients with lung cancer allows the rapid identification of a broad array of targetable molecular drivers. We hypothesized that...
- Published
- 2018
33. Tumor mutation burden and efficacy of targeted therapy in patients with EGFR mutant lung cancers
- Author
-
Megan Tenet, Mark G. Kris, Hira Rizvi, Gregory J. Riely, Helena Alexandra Yu, Ai Ni, Matthew D. Hellmann, Francisco Sanchez Vega, Charles M. Rudin, and Michael Offin
- Subjects
Cancer Research ,Lung ,business.industry ,medicine.medical_treatment ,Mutant ,Immune checkpoint ,Targeted therapy ,Blockade ,medicine.anatomical_structure ,Oncology ,Mutation (genetic algorithm) ,Cancer research ,medicine ,Biomarker (medicine) ,In patient ,business - Abstract
9042Background: Tumor mutation burden (TMB) is a biomarker of response to immune checkpoint blockade. The impact of TMB on outcomes with targeted therapies has not been explored. We hypothesized th...
- Published
- 2018
34. Frequency of brain metastases and outcomes in patients with HER2-, KRAS-, and EGFR-mutant lung cancers
- Author
-
Lisa M. DeAngelis, James M. Isbell, Wei-Chu Victoria Lai, Alexander Drilon, Ai Ni, Kathryn Beal, Robert J. Young, Bob T. Li, Mark G. Kris, David R. Jones, Maria E. Arcila, Daniel Feldman, Gregory J. Riely, Helena Alexandra Yu, Michael Offin, Elena Pentsova, Kathryn C. Arbour, Mariza Daras, and Emmet Jordan
- Subjects
Cancer Research ,Lung ,business.industry ,Mutant ,respiratory system ,medicine.disease_cause ,respiratory tract diseases ,medicine.anatomical_structure ,Oncology ,Cancer research ,medicine ,In patient ,KRAS ,skin and connective tissue diseases ,business ,neoplasms - Abstract
9081Background: HER2 mutations are oncogenic drivers in 3% of lung cancers. Here we describe the frequency and course of patients with HER2 mutant lung cancers with spread to brain and compare resu...
- Published
- 2018
35. Circulating tumor DNA in advanced lung cancers: A prospective evaluation of matched therapy and shedding detection
- Author
-
Joshua K. Sabari, David R. Jones, Ai Ni, Stephen Clarke, Mark Li, James M. Isbell, Nick Pavlakis, Sutirtha Datta, Charles M. Rudin, Maria E. Arcila, Dennis Stephens, Andreas Rimner, Alexander Drilon, Alex Makhnin, Valerie W. Rusch, Nidhi Tandon, Bob T. Li, Michael Offin, Mackenzie L. Myers, and Lee Lim
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,business.industry ,medicine.disease ,DNA sequencing ,Prospective evaluation ,Clinical Practice ,medicine.anatomical_structure ,Circulating tumor DNA ,Internal medicine ,medicine ,Liquid biopsy ,Lung cancer ,business - Abstract
e21234Background: Liquid biopsy of circulating tumor DNA (ctDNA) by next generation sequencing (NGS) is widely adopted in clinical practice. ctDNA shedding across different lung cancer histologies,...
- Published
- 2018
36. RET-rearranged lung cancers: Immunophenotype and response to immunotherapy
- Author
-
Kathryn C. Arbour, Matthew D. Hellmann, Darragh Halpenny, Ai Ni, Gregory J. Riely, Dazhi Liu, Mark G. Kris, Wei-Chu Victoria Lai, Natasha Rekhtman, Josiah D. Land, Stephanie L. Wu, Joshua K. Sabari, Alexander Drilon, Michael Offin, Charles M. Rudin, Bob T. Li, Terry K. Pak, and Joseph Montecalvo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Cabozantinib ,medicine.medical_treatment ,Vandetanib ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immunophenotyping ,Internal medicine ,Medicine ,Lung cancer ,Chemotherapy ,business.industry ,Immunotherapy ,medicine.disease ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Immunohistochemistry ,business ,medicine.drug - Abstract
9034Background: In patients with RET-rearranged lung cancers, multikinase inhibitors (e.g. cabozantinib and vandetanib) and specific (e.g. pemetrexed-containing) chemotherapy regimens have documented activity. In contrast, PD-L1 expression, tumor mutational burden (TMB), and the treatment outcomes of immunotherapy are not well characterized in this genomic subset. Methods: Patients with a pathologically confirmed diagnosis of lung cancer harboring a RET rearrangement were identified between January 2012 and December 2017. PD-L1 expression was determined by IHC (E1L3N Clone). TMB was calculated in tumors that underwent NGS (Mutations/Mb, MSK-IMPACT). Objective response rate (ORR) to immunotherapy was evaluated by RECIST. Results: 74 patients with RET-rearranged lung cancers were identified. The median age was 63 (range 34-80 years), 55% were women, and 68% were never smokers. The majority (97%) had adenocarcinoma and 85% had metastatic disease. In patients with sufficient tissue for analysis, tumor PD-L1 e...
- Published
- 2018
37. Outcomes of advanced pulmonary large cell neuroendocrine carcinoma stratified by RB1 loss, SLFN11 expression, and tumor mutational burden
- Author
-
Charles M. Rudin, Darragh Halpenny, Ai Ni, Matthew D. Hellmann, Alexander Drilon, Ricklie Ann Julian, Joshua K. Sabari, Bob T. Li, John T. Poirier, and Natasha Rekhtman
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Disease ,Large cell neuroendocrine carcinoma of the lung ,medicine.disease ,eye diseases ,respiratory tract diseases ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,business ,neoplasms - Abstract
e20568Background: LCNEC is a biologically heterogeneous disease comprised of distinct subgroups characterized by genomic signatures of either SCLC or NSCLC (e.g. RB1 loss or retained). We assessed ...
- Published
- 2018
38. PD-L1 expression, tumor mutation burden and response to immune checkpoint blockade in patients with HER2-mutant lung cancers
- Author
-
Andrew J. Plodkowski, Matthew D. Hellmann, Wei-Chu Victoria Lai, Ai Ni, Edyta B. Brzostowski, Joshua K. Sabari, Alexander Drilon, Maria E. Arcila, Daniel Feldman, Mark G. Kris, Hira Rizvi, Michael Offin, Darren J. Buonocore, and Bob T. Li
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,Lung ,business.industry ,Mutant ,respiratory system ,medicine.disease_cause ,Immune checkpoint ,respiratory tract diseases ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,In patient ,Pd l1 expression ,business - Abstract
9060Background: HER2 mutations are present in 3% of lung cancers. Response to immune checkpoint blockade (ICB) in this subset of lung cancers is unknown. We evaluate the landscape of PD-L1 and tumo...
- Published
- 2018
39. Deleterious effect of baseline steroids on efficacy of PD-(L)1 blockade in patients with NSCLC
- Author
-
Joshua K. Sabari, Darragh Halpenny, Roberto Ferrara, Ai Ni, Jamie E. Chaft, Andrew J. Plodkowski, Benjamin Besse, Gala Martinez Bernal, Caroline Caramella, Lizza E.L. Hendriks, Niamh Long, Wei-Chu Victoria Lai, Kathryn C. Arbour, Matthew D. Hellmann, Edouard Auclin, Gregory J. Riely, Laura Mezquita, Hira Rizvi, and David Planchard
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.disease ,Blockade ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,Lung cancer ,business ,Standard therapy ,Immunosuppressive effect - Abstract
9003Background: Treatment with PD-(L)1 inhibitors is now standard therapy for patients with lung cancer. The immunosuppressive effect of steroids may reduce efficacy of PD-(L)1 blockade. On-treatme...
- Published
- 2018
40. Refining actionable HER2 alterations in lung cancers through next generation sequencing (NGS)
- Author
-
Pedram Razavi, Ai Ni, Wei-Chu Victoria Lai, Edyta B. Brzostowski, Maria E. Arcila, Lillian M. Smyth, Michael F. Berger, Marc Ladanyi, Chongrui Xu, Charles M. Rudin, Bob T. Li, Alexander Drilon, Mark G. Kris, Matthew D. Hellmann, Darren J. Buonocore, Tao Zheng, David B. Solit, Ronglai Shen, Mackenzie L. Myers, and Ahmet Zehir
- Subjects
Cancer Research ,Lung ,medicine.anatomical_structure ,Oncology ,Drug development ,business.industry ,Mutation (genetic algorithm) ,Medicine ,Computational biology ,business ,DNA sequencing ,Protein overexpression - Abstract
e24181Background: Targeting HER2 alterations including mutation, amplification and protein overexpression has been an active area of new drug development. However, the relationships between these H...
- Published
- 2018
41. A phase II study of lenvatinib in patients with progressive, recurrent/metastatic adenoid cystic carcinoma
- Author
-
Vatche Tchekmedyian, Eric Jeffrey Sherman, Lara Dunn, Crystal Tran, Nora Katabi, Ai Ni, Sofia Haque, David G. Pfister, and Alan Loh Ho
- Subjects
Cancer Research ,Oncology - Published
- 2018
42. Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer
- Author
-
Matthew D. Hellmann, Hossein Borghaei, Megan Tenet, Francisco Sanchez-Vega, Jeff Hammerbacher, Patrik Vitazka, Margaret K. Callahan, Jamie E. Chaft, Cailian Liu, Jennifer L. Sauter, William J. Geese, Arun Ahuja, Levi Mangarin, Taha Merghoub, Tavi Nathanson, Jacqueline Buros Novik, Nicholas McGranahan, Kelly L. Covello, Scott J. Antonia, Charles Swanton, Natasha Rekhtman, Andrea Renninger, Ai Ni, Mohsen Abu-Akeel, Alexandra Snyder, Martin H. Voss, Eliza Chang, Xuemei Li, Hira Rizvi, Jedd D. Wolchok, Benjamin C. Creelan, and Charles M. Rudin
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Combination therapy ,medicine.medical_treatment ,Ipilimumab ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Progression-free survival ,Lung cancer ,business.industry ,Immunotherapy ,medicine.disease ,Immune checkpoint ,respiratory tract diseases ,3. Good health ,Blockade ,030104 developmental biology ,030220 oncology & carcinogenesis ,Nivolumab ,business ,medicine.drug - Abstract
Combination immune checkpoint blockade has demonstrated promising benefit in lung cancer, but predictors of response to combination therapy are unknown. Using whole-exome sequencing to examine non-small-cell lung cancer (NSCLC) treated with PD-1 plus CTLA-4 blockade, we found that high tumor mutation burden (TMB) predicted improved objective response, durable benefit, and progression-free survival. TMB was independent of PD-L1 expression and the strongest feature associated with efficacy in multivariable analysis. The low response rate in TMB low NSCLCs demonstrates that combination immunotherapy does not overcome the negative predictive impact of low TMB. This study demonstrates the association between TMB and benefit to combination immunotherapy in NSCLC. TMB should be incorporated in future trials examining PD-(L)1 with CTLA-4 blockade in NSCLC.
- Published
- 2018
43. Germline Variants in Targeted Tumor Sequencing Using Matched Normal DNA
- Author
-
Kasmintan A. Schrader, Meera Prasad, Tinu Thomas, Asad Ashraf, Annie Lincoln, Ahmet Zehir, Mark E. Robson, Ai Ni, Liying Zhang, Maria E. Arcila, David S. Klimstra, Ryma Benayed, Kenneth Offit, Donavan T. Cheng, David B. Solit, David M. Hyman, Zsofia K. Stadler, Michael F. Berger, Vijai Joseph, Marc Ladanyi, and Michael Walsh
- Subjects
0301 basic medicine ,Adult ,Male ,Cancer Research ,DNA Mutational Analysis ,Bioinformatics ,Risk Assessment ,Article ,DNA sequencing ,Germline ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Germline mutation ,Predictive Value of Tests ,Risk Factors ,Neoplasms ,Biomarkers, Tumor ,Medicine ,Humans ,Genetic Predisposition to Disease ,Allele ,Precision Medicine ,Gene ,Germ-Line Mutation ,Aged ,Aged, 80 and over ,Models, Genetic ,business.industry ,Gene Expression Profiling ,Middle Aged ,Prognosis ,Phenotype ,Gene expression profiling ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Mendelian inheritance ,symbols ,Female ,New York City ,business - Abstract
Importance Tumor genetic sequencing identifies potentially targetable genetic alterations with therapeutic implications. Analysis has concentrated on detecting tumor-specific variants, but recognition of germline variants may prove valuable as well. Objective To estimate the burden of germline variants identified through routine clinical tumor sequencing. Design, Setting, and Participants Patients with advanced cancer diagnoses eligible for studies of targeted agents at Memorial Sloan Kettering Cancer Center are offered tumor-normal sequencing with MSK-IMPACT, a 341-gene panel. We surveyed the germline variants seen in 187 overlapping genes with Mendelian disease associations in 1566 patients who had undergone tumor profiling between March and October 2014. Main Outcomes and Measures The number of presumed pathogenic germline variants (PPGVs) and variants of uncertain significance per person in 187 genes associated with single-gene disorders and the proportions of individuals with PPGVs in clinically relevant gene subsets, in genes consistent with known tumor phenotypes, and in genes with evidence of second somatic hits in their tumors. Results The mean age of the 1566 patients was 58 years, and 54% were women. Presumed pathogenic germline variants in known Mendelian disease-associated genes were identified in 246 of 1566 patients (15.7%; 95% CI, 14.0%-17.6%), including 198 individuals with mutations in genes associated with cancer susceptibility. Germline findings in cancer susceptibility genes were concordant with the individual’s cancer type in only 81 of 198 cases (40.9%; 95% CI, 34.3%-47.9%). In individuals with PPGVs retained in the tumor, somatic alteration of the other allele was seen in 39 of 182 cases (21.4%; 95% CI, 16.1%-28.0%), of which 13 cases did not show a known correlation of the germline mutation and a known syndrome. Mutations in non–cancer-related Mendelian disease genes were seen in 55 of 1566 cases (3.5%; 95% CI, 27.1%-45.4%). Almost every individual had more than 1 variant of uncertain significance (1565 of 1566 patients; 99.9%; 95% CI, 99.6%-99.9%). Conclusions and Relevance Germline variants are common in individuals undergoing tumor-normal sequencing and may reveal otherwise unsuspected syndromic associations.
- Published
- 2015
44. Genetic testing decision-making experiences within families of colorectal cancer patients: A qualitative study.
- Author
-
He, Meng, Guan, Jingjing, Li, Huanhuan, Dong, Yueyang, Zhu, Xiangning, Sun, Juanjuan, Gao, Shizheng, Zhi, Shengze, Ai ni, Bu he li qian mu, and Sun, Jiao
- Abstract
Genetic testing is the gold standard for the diagnosis of hereditary colorectal cancer syndromes but is currently inadequate and nonideal. The decision-making processes regarding genetic testing are even less well known. The present study aims to explore the decision-making experience of genetic testing for colorectal cancer patients and their family members. A descriptive qualitative study was employed. Data were collected using individual semi-structured interviews with 5 colorectal cancer patients and 20 family members from November 2020 to April 2021. Interviews were transcribed and analysed using inductive content analysis. Four categories were identified: 1) the source of information for genetic testing, 2) the differentiated attitudes towards genetic testing, 3) genetic testing decisional needs, and 4) the factors influencing genetic testing decision-making. Colorectal cancer patients and their families engaged in two distinct pathways to genetic testing decisions: direct decision-making and indirect decision-making. Throughout these processes, due to the limited source of information, they had information needs that were met and facilitated genetic testing decision-making. Colorectal cancer patients and family members need knowledge related to genetic testing, but they have limited access to information, which prevents them from making informed decisions. Providing decision aid interventions and informational support are significant steps towards addressing the support needs of this population. • Genetic testing is important, but is currently inadequate and nonideal. • There is currently limited understanding of genetic testing decision-making process. • Two distinct decision-making paths are generated regarding genetic testing. • The family plays an important role in genetic testing decision-making process. • Colorectal cancer patients and their families have decisional needs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
45. Lung cancers with mutations in EGFR exon 18: Molecular characterization and clinical outcomes in response to tyrosine kinase inhibitors
- Author
-
Wei-Chu Victoria Lai, Gregory J. Riely, Maria E. Arcila, Joshua K. Sabari, Ai Ni, Kathryn C. Arbour, Helena Alexandra Yu, Charles M. Rudin, Mark G. Kris, and James Huang
- Subjects
Cancer Research ,Exon ,Lung ,medicine.anatomical_structure ,Oncology ,business.industry ,medicine ,Cancer research ,business ,Tyrosine kinase - Abstract
9029 Background: Little data is available to guide clinical management of individuals with less common oncogenic drivers such as exon 18 mutations (ex18m) in EGFR. To better understand the impact of these rare mutations on treatment outcomes, we reviewed clinicopathologic data in patients (pts) with ex18m treated with tyrosine kinase inhibitors (TKI) in EGFR-mutant lung cancers. Methods: Pts with EGFR ex18m were detected via molecular diagnostics using Sequenom™, FoundationOne™ or MSK IMPACT™ NGS testing from 2003-2016. We reviewed their clinical data for molecular alterations in EGFR, treatment outcomes in response to TKI (time on treatment) and median overall survival (OS). Results: We identified mutations in EGFR ex18m in 63 pts. Median age at diagnosis was 68; 63% were women; 29% never smokers. Overall, 74 ex18m were found in 63 pts, including: G719A = 38, G719S = 11, G719C = 8, E709K = 6, E709_T710delinsD = 6, E709A = 3, G719D = 2. E709 and G719 co-mutations in ex18 were found in 9 pts, and 1 pt was found to have 3 separate tumors, each with a distinct ex18m. 29/63 (46%) patients with ex18m had a co-occurring EGFR mutation: 9 with another ex18m; 20 with ex19-21m. Using our IMPACT NGS, the median number of co-mutations was 8 (range 1-17). Two out of 63 pts had a pre-treatment T790M mutation. The 25 pts with non-metastatic disease presented in the following stages: IA = 19; IB = 3; IIB = 1; IIIA = 2; IIIB = 2. 34/38 pts with metastatic disease were treated with the following first-line EGFR-TKIs: erlotinib = 28, afatinib = 5, osimertinib = 1. Median duration on TKI treatment in months was: erlotinib = 10 mo, (range 1-25), afatinib = 3 mo (range 2-9), osimertinib = 4 mo. Median OS from the date of diagnosis of metastatic disease was 22 months (95% CI 18-29). In comparison, a similar cohort of pts with sensitizing EGFR exon19del/L858R mutations had a median OS of 31 months (95% CI 28-33) (Naidoo Cancer2015). Conclusions: Almost half of ex18m occur concurrently with another EGFR mutation. Overall, ex18m pts have a shorter median OS when compared to similar patient cohorts. EGFR-TKIs appear to be an effective treatment for pts with ex18m in EGFR-mutant lung cancers.
- Published
- 2017
46. Afatinib in patients with metastatic HER2-mutant lung cancers: An international multicenter study
- Author
-
Ai Ni, Charles M. Rudin, Maria E. Arcila, Nick Pavlakis, Tristan A. Barnes, Julie Milia, Roberto Ferrara, Joshua K. Sabari, Bob T. Li, Oliver Gautschi, Julien Mazieres, Mark G. Kris, Natasha B. Leighl, Louisiane Lebas, Frances A. Shepherd, Wei-Chu Victoria Lai, Solange Peters, and Stephen Clarke
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,medicine.drug_class ,Human epidermal growth factor ,business.industry ,Afatinib ,Mutant ,Tyrosine-kinase inhibitor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Multicenter study ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,business ,medicine.drug - Abstract
9071 Background: Human epidermal growth factor 2 ( HER2, ERBB2) mutations have been identified as oncogenic drivers in 3% of lung cancers. Afatinib is an irreversible tyrosine kinase inhibitor of HER1 (EGFR), HER2 and HER4 and has been described in case reports to have activity in HER2-mutant lung cancers. However, there is little data to inform the clinical use of afatinib. Methods: We reviewed patients with metastatic HER2-mutant lung cancers treated with afatinib among 7 institutions between 2009 and 2016. The primary endpoint was investigator assessed overall response rate using RECIST v1.1. Other data collected included types of HER2mutations, duration of afatinib treatment and overall survival. Results: We identified 27 patients with metastatic HER2-mutant lung cancers treated with afatinib. Median age at diagnosis was 63 (range 40 to 84); majority were men (n = 16; 59%) and never-smokers (n = 18; 67%). All tumors were adenocarcinomas, and the majority were Stage IV at initial diagnosis (n = 16; 59%). A 12-base pair (bp) in-frame insertion YVMA in exon 20 (p.A775_G776insYVMA) was present in 16 patients (59%). In addition, there were three 9-bp insertions, two 3-bp insertions and two single bp substitutions (L755F and D769H) in exon 20; two single bp substitutions (S310F) in exon 8; one exon 17 V659E mutation; and one single-nucleotide polymorphism (Ile655Val). Median duration on afatinib was 2 months (range 1 to 27); median line of prior treatment was 3 (range 1 to 6). Eight patients had previously received trastuzumab prior to afatinib and one concurrently with afatinib. Overall response rate was 15% (n = 4; 95% CI 4 to 34%); the four partial responses lasted 5, 5, 6 and 10 months. The 3 longest partial responders had a 12-bp insertion in exon 20 (YVMA); the remaining partial responder had a 9-bp insertion in exon 20. Median overall survival from diagnosis date of metastatic disease was 23 months (95% CI 18 to 62). Conclusions: Afatinib produced partial responses in 15% of patients with metastatic HER2-mutant lung cancers, including insertion YVMA. Our findings confirm the activity of afatinib and provide data supporting a framework for its use in the care of patients with HER2-mutant lung cancers.
- Published
- 2017
47. Response to osimertinib following treatment with EGF816 in patients with T790M EGFR mutant NSLCLC
- Author
-
Paul K. Paik, Ai Ni, Helena Alexandra Yu, Mark G. Kris, Zofia Piotrowska, Kathryn C. Arbour, Gregory J. Riely, Lecia V. Sequist, and Andrew J. Plodkowski
- Subjects
0301 basic medicine ,Cancer Research ,business.industry ,Mutant ,Pharmacology ,Third generation ,respiratory tract diseases ,03 medical and health sciences ,T790M ,Egfr tki ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Epidermal growth factor ,030220 oncology & carcinogenesis ,Medicine ,In patient ,Osimertinib ,business ,Tyrosine kinase - Abstract
e20673 Background: Third generation (3rd gen) epidermal growth factor (EGFR) tyrosine kinase inhibitors (TKIs) have been developed to treat EGFR T790M-mediated resistance to EGFR TKIs by inhibiting EGFR T790M, as well as EGFR L858R and EGFR exon 19 deletions. The mechanisms of resistance to third-generation EGFR TKIs are largely unknown and clinical cross-resistance among 3rd gen EGFR TKIs has not been routinely evaluated. Osimertinib is an FDA-approved irreversible 3rd gen EGFR TKI. In patients with EGFR T790M mutant NSCLC, the response rate (ORR) to osimertinib is 61%. EGF816 is a covalent, irreversible, 3rd gen EGFR TKI in clinical development. In early phase data of EGF816, the ORR was 47% and disease control rate was 87% in patients with EGFR T790M mutant NSCLC. To assess clinical cross-resistance between EGF816 and osimertinib, we evaluated the clinical outcomes of patients treated with osimertinib in patients previously treated with EGF816 during the phase I/II trial. Methods: Patients with metastatic EGFR mutant lung adenocarcinoma were identified who were previously treated with EGF816 and received osimertinib after progression of disease on EGF816 (NCT02108964). All patients had documented T790M mutation prior to treatment with EGF816. The best overall response to osimertinib was determined by RECIST 1.1 criteria. Duration of clinical benefit was defined as duration of osimertinib therapy. Results: Fourteen (3 men, 11 women, median age 58 [range 33-77]) patients met eligibility criteria at our centers. The ORR to subsequent osimertinib therapy was 14% (1 CR, 1 PR, 8 SD, 4 POD). Patients continued treatment with osimertinib for a median of 9 months (95% CI 3.8-10.1, [median follow up 11 months, range 1-13 months]). 5 patients are still on osimertinib to date (one patient each 3+, 6+, 8+, 11+, and 12+ months). Conclusions: This series suggests a potentially meaningful clinical benefit for patients with sequential therapy with two different third-generation EGFR inhibitors, emphasizing the importance of understanding resistance mechanisms (genetic alteration of target, bypass signaling, pharmacology, etc.) and raising the possibility of the need for multiple third generation EGFR TKIs in clinical practice.
- Published
- 2017
48. Pulse-continuous dose erlotinib as initial targeted therapy for patients with EGFR-mutant lung cancers with untreated brain metastases
- Author
-
Kathryn Beal, Christopher R. Rodriguez, Ai Ni, Kathryn C. Arbour, Robert J. Young, Gregory J. Riely, Sara A. Hayes, Helena Alexandra Yu, Mariza Daras, Mark G. Kris, and W. Pao
- Subjects
Cancer Research ,Lung ,Pulse (signal processing) ,business.industry ,medicine.medical_treatment ,Mutant ,Targeted therapy ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Erlotinib ,business ,medicine.drug - Abstract
9039 Background: Clarke (Neurooncol 2010) reported responses with intermittent high pulse doses of erlotinib (leading to higher concentrations in CSF) given to patients with EGFR-mutant central nervous system metastases developing on standard erlotinib doses. In a phase 1 study of pulse-continuous dose erlotinib, no patient developed progression in existing or new brain or leptomeningeal metastases (Yu Ann Oncol 2016). This phase 2 trial tested pulse-continuous dose erlotinib in patients with lung cancers with EGFRmutations with brain metastases. Methods: Patients had no prior EGFR TKI or radiation to the brain and at least 1 target brain metastasis. All received initial daily "pulse" doses of erlotinib 1200 mg days 1&2 and "continuous" 50 mg doses days 3-7 (doses and schedule from the Yu Phase 1 study), weekly until progression. The co-primary endpoints were overall and brain metastasis response by RECIST 1.1. Results: We enrolled 19 patients with EGFR-mutant lung cancers: median age 61yrs (range 45-80), 74% women, 95% Karnofsky PS ≥80%, 1 leptomeningeal disease, 33% prior pemetrexed-based chemotherapy. The median size of target brain metastases was 13 mm (range 10-19 mm). 32% were on dexamethasone for cerebral edema. The partial response rate overall was 74% (95% CI 51-89%) and also 74% in brain metastases. Of 10 patients with progression, 9/10 occurred in non-brain sites (4 EGFRT790M, 1 with progression in brain as well), 1 with leptomeningeal. The median progression free survival was 10 mo (range 7-NR mo). Pulse doses were reduced in 68% (median delivered pulse dose 1050 mg days 1&2, range 600-1200 mg). Incidences of any gradeof rash and diarrhea were 84% and 63% respectively. There were no grade 4 or 5 toxicities. Conclusions: Pulse-continuous dose erlotinib alone controlled brain and leptomeningeal metastases in 89% (95% CI 67-98%) of patients with EGFR-mutant lung cancers with central nervous system spread pretreatment, with an overall response rate of 74% and progression free survival and rates of rash and diarrhea comparable to series with erlotinib 150 mg daily. Supported by Astellas, CA 129243, CA 008748. NCT01967095 Clinical trial information: NCT01967095.
- Published
- 2017
49. Liquid biopsy in the clinic: A prospective study of plasma circulating tumor DNA (ctDNA) next generation sequencing (NGS) in patients with advanced non-small cell lung cancers to match targeted therapy
- Author
-
Charles M. Rudin, Joshua K. Sabari, Ai Ni, Gregory J. Riely, Nidhi Tandon, Samantha Henderson, David R. Jones, Sutirtha Datta, Lee Lim, Christopher K. Raymond, Bob T. Li, Adrian Lee, Valerie W. Rusch, Michael Offin, Andres Martinez, James M. Isbell, Mark Li, Nick Pavlakis, Stephen Clarke, and Mariel A. DuBoff
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung ,business.industry ,medicine.medical_treatment ,Concordance ,Bioinformatics ,DNA sequencing ,Targeted therapy ,medicine.anatomical_structure ,Internal medicine ,medicine ,Clinical endpoint ,In patient ,Liquid biopsy ,Prospective cohort study ,business - Abstract
11536 Background: Liquid biopsy for plasma ctDNA NGS is a rapidly evolving science. Plasma ctDNA assays are commercially available and are increasingly adopted in the community with a paucity of evidence-based guidance. We set out to study the optimal timing and utility of plasma ctDNA NGS in clinic. Methods: Pts with advanced NSCLC who were driver unknown, defined as not having prior tissue NGS or clinical concern for tumor heterogeneity that may affect treatment decisions, were eligible. Peripheral blood was collected in a Streck tube (10mL), DNA extracted, and subjected to a bias-corrected hybrid-capture 21 gene targeted NGS assay in a CLIA lab with unique reads at 3000x and sensitive detection at variant allele frequency above 0.1% (ResolutionBio Bellevue, WA). Pts also had concurrent tissue NGS via MSK IMPACT. Clinical endpoints included detection of oncogenic drivers in plasma, ability to match pts to targeted therapy, concordance and turnaround time of plasma and tissue NGS. Results: Forty-one pts were prospectively accrued. Plasma ctDNA detected an oncogenic driver in 39% (16/41) of pts, of whom 17% (7/41) were matched to targeted therapy; including pts matched to clinical trials for HER2 exon 20 insertionYVMA, BRAF L597Q and MET exon14. Mean turnaround time for plasma was 7 days (4-12) and 28 days (20-43) for tissue. Plasma ctDNA was detected in 56% (23/41) of pts; detection was 40% (8/20) if blood was drawn on active therapy and 71% (15/21) if drawn off therapy, either at diagnosis or progression (Odds ratio 0.28, 95% CI 0.06 - 1.16; p = 0.06). All pts had concurrent tissue NGS; of the 10 samples resulted, there was 100% driver concordance between tissue and plasma in pts drawn off therapy. Conclusions: In pts who were driver unknown or who had clinical concern for tumor heterogeneity, plasma ctDNA NGS identified a variety of oncogenic drivers with a short turnaround time and matched them to targeted therapy. Plasma ctDNA detection was more frequent at diagnosis of metastatic disease or at progression. A positive finding of an oncogenic driver in plasma is highly specific, but a negative finding may still require tissue biopsy.
- Published
- 2017
50. Phase I dose escalation of ibrutinib and buparlisib in relapsed/refractory diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), and follicular lymphoma (FL)
- Author
-
Heiko Schöder, Thu Oanh Dang, David J. Straus, Salma Ahsanuddin, Connie Lee Batlevi, Ahmet Dogan, Paul A. Hamlin, Craig H. Moskowitz, John F. Gerecitano, Jürgen Rademaker, Ai Ni, Amanda R Copeland, Benjamin Freidin, Anas Younes, Pamela Drullinsky, Steven M. Horwitz, Matthew J. Matasar, Andrew D. Zelenetz, Devin Callan, and Robert Porzio
- Subjects
0301 basic medicine ,Cancer Research ,biology ,business.industry ,Buparlisib ,Follicular lymphoma ,medicine.disease ,Lymphoma ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Oncology ,chemistry ,hemic and lymphatic diseases ,Ibrutinib ,biology.protein ,medicine ,Dose escalation ,Cancer research ,Bruton's tyrosine kinase ,Mantle cell lymphoma ,business ,Diffuse large B-cell lymphoma - Abstract
7544 Background: In vitro studies of BTK and PI3K inhibitors demonstrate synergy in non-Hodgkin lymphoma (NHL). We embarked on a phase I/Ib investigator-initiated clinical trial evaluating the combination of ibrutinib (BTK inhibitor) and buparlisib (pan-PI3K inhibitor) in relapsed/refractory (R/R) NHL. The completed dose escalation is reported. Methods: Patients (pts) were eligible if they had R/R DLBCL, MCL, or FL with ECOG≤2 and adequate organ function. Ibrutinib and buparlisib were given daily by mouth on a 28-day cycle. Dose reductions were permitted after cycle 1. Tumor response was based on Lugano Classification however CR required both PET resolution and ≥ PR by CT. Results: As of Dec 16, 2016, 13 pts were enrolled and evaluated for toxicity (DLBCL 5, FL 2, MCL 6). Dose levels and DLT per table. Six pts discontinued treatment for disease progression (DLBCL 4, FL 2). Hematologic AE ≥ grade 3 are anemia (2), leukocytosis (2), and leukopenia (4). Relevant non-hematologic AEs of any grade ≥ 20% across all pts were fatigue (77%), diarrhea (62%), anorexia (54%), rash (46%), hyperbilirubinemia (46%), gastric reflux (46%), CMV reactivation (31%), mood change (31%), and hypertension (23%). Most common related grade 3/4 toxicity is rash (N = 3). No grade 5 toxicities noted. Serious adverse events (SAE) include: grade 2 pleural effusion and grade 2 nausea (N = 1), grade 1 fever with hospitalization (N = 1), grade 2 confusion and grade 4 hyponatremia (N = 1) were unrelated to therapy. Responses noted in 13 pts: MCL (N = 6: CR 4, PR 2), FL (N = 2: SD 2), DLBCL (N = 5: SD 1). One CR was a MCL pt with CR after 2 cycles on combination therapy and continues in remission on ibrutinib alone because of buparlisib toxicity. Conclusions: Combination of ibrutinib and buparlisib while generally well tolerated has predicted toxicities of both BTK and PI3K inhibitors. The recommended phase 2 dose is ibrutinib 560 mg and buparlisib 100 mg though dose reductions for tolerability may be needed for long term oral therapies. Promising efficacy is observed in MCL. Clinical trial information: NCT02756247. [Table: see text]
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.