40 results on '"Baik CS"'
Search Results
2. Emerging molecular biomarkers--blood-based strategies to detect and monitor cancer.
- Author
-
Hanash SM, Baik CS, Kallioniemi O, Hanash, Samir M, Baik, Christina S, and Kallioniemi, Olli
- Abstract
There is an urgent need for blood-based, noninvasive molecular tests to assist in the detection and diagnosis of cancers in a cost-effective manner at an early stage, when curative interventions are still possible. Additionally, blood-based diagnostics can classify tumors into distinct molecular subtypes and monitor disease relapse and response to treatment. Increasingly, biomarker strategies are becoming critical to identify a specific patient subpopulation that is likely to respond to a new therapeutic agent. The improved understanding of the underlying molecular features of common cancers and the availability of a multitude of recently developed technologies to interrogate the genome, transcriptome, proteome and metabolome of tumors and biological fluids have made it possible to develop clinically applicable and cost-effective tests for many common cancers. Overall, the paradigm shift towards personalized and individualized medicine relies heavily on the increased use of diagnostic biomarkers and classifiers to improve diagnosis, management and treatment. International collaborations, involving both the private and public sector will be required to facilitate the development of clinical applications of biomarkers, using rigorous standardized assays. Here, we review the recent technological and scientific advances in this field. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
3. EGFR Mutations in Squamous Cell Lung Cancer in Never-Smokers.
- Author
-
Baik CS, Pritchard CC, Eaton KD, and Chow LQ
- Published
- 2013
- Full Text
- View/download PDF
4. Safety and efficacy of pralsetinib in RET fusion–positive non-small-cell lung cancer including as first-line therapy
- Author
-
F. Griesinger, G. Curigliano, M. Thomas, V. Subbiah, C.S. Baik, D.S.W. Tan, D.H. Lee, D. Misch, E. Garralda, D.-W. Kim, A.J. van der Wekken, J.F. Gainor, L. Paz-Ares, S.V. Liu, G.P. Kalemkerian, Y. Houvras, D.W. Bowles, A.S. Mansfield, J.J. Lin, V. Smoljanovic, A. Rahman, S. Kong, A. Zalutskaya, M. Louie-Gao, A.L. Boral, J. Mazières, Institut Català de la Salut, [Griesinger F] Department of Hematology and Oncology, Pius-Hospital, University of Oldenburg, Oldenburg, Germany. [Curigliano G] European Institute of Oncology, IRCCS, Milan, Italy. Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy. [Thomas M] Department of Thoracic Oncology, Thoraxklinik, University Heidelberg and Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany. [Subbiah V] Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, USA. [Baik CS] Division of Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, USA. [Tan DSW] Division of Medical Oncology, National Cancer Centre Singapore, Singapore. [Garralda E] Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
Otros calificadores::/uso terapéutico [Otros calificadores] ,Medicaments antineoplàstics - Ús terapèutic ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,acciones y usos químicos::acciones farmacológicas::usos terapéuticos::antineoplásicos [COMPUESTOS QUÍMICOS Y DROGAS] ,diagnóstico::pronóstico::resultado del tratamiento [TÉCNICAS Y EQUIPOS ANALÍTICOS, DIAGNÓSTICOS Y TERAPÉUTICOS] ,Hematology ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,Diagnosis::Prognosis::Treatment Outcome [ANALYTICAL, DIAGNOSTIC AND THERAPEUTIC TECHNIQUES, AND EQUIPMENT] ,targeted therapy ,Oncology ,non-small-cell lung cancer ,pralsetinib ,Avaluació de resultats (Assistència sanitària) ,Other subheadings::/therapeutic use [Other subheadings] ,Chemical Actions and Uses::Pharmacologic Actions::Therapeutic Uses::Antineoplastic Agents [CHEMICALS AND DRUGS] ,Pulmons - Càncer - Tractament ,frontline therapy ,RET fusion ,RET inhibition - Abstract
RET inhibition; Pralsetinib; Targeted therapy Inhibición de RET; Pralsetinib; Terapia dirigida Inhibició de RET; Pralsetinib; Teràpia dirigida Background RET fusions are present in 1%-2% of non-small-cell lung cancer (NSCLC). Pralsetinib, a highly potent, oral, central nervous system-penetrant, selective RET inhibitor, previously demonstrated clinical activity in patients with RET fusion–positive NSCLC in the phase I/II ARROW study, including among treatment-naive patients. We report an updated analysis from the ARROW study. Patients and methods ARROW is a multi-cohort, open-label, phase I/II study. Eligible patients were ≥18 years of age with locally advanced or metastatic solid tumours and an Eastern Cooperative Oncology Group performance status of 0-2 (later 0-1). Patients initiated pralsetinib at the recommended phase II dose of 400 mg once daily until disease progression, intolerance, consent withdrawal, or investigator’s decision. The co-primary endpoints (phase II) were overall response rate (ORR) by blinded independent central review and safety. Results Between 17 March 2017 and 6 November 2020 (data cut-off), 281 patients with RET fusion–positive NSCLC were enrolled. The ORR was 72% [54/75; 95% confidence interval (CI) 60% to 82%] for treatment-naive patients and 59% (80/136; 95% CI 50% to 67%) for patients with prior platinum-based chemotherapy (enrolment cut-off for efficacy analysis: 22 May 2020); median duration of response was not reached for treatment-naive patients and 22.3 months for prior platinum-based chemotherapy patients. Tumour shrinkage was observed in all treatment-naive patients and in 97% of patients with prior platinum-based chemotherapy; median progression-free survival was 13.0 and 16.5 months, respectively. In patients with measurable intracranial metastases, the intracranial response rate was 70% (7/10; 95% CI 35% to 93%); all had received prior systemic treatment. In treatment-naive patients with RET fusion–positive NSCLC who initiated pralsetinib by the data cut-off (n = 116), the most common grade 3-4 treatment-related adverse events (TRAEs) were neutropenia (18%), hypertension (10%), increased blood creatine phosphokinase (9%), and lymphopenia (9%). Overall, 7% (20/281) discontinued due to TRAEs. Conclusions Pralsetinib treatment produced robust efficacy and was generally well tolerated in treatment-naive patients with advanced RET fusion–positive NSCLC. Results from the confirmatory phase III AcceleRET Lung study (NCT04222972) of pralsetinib versus standard of care in the first-line setting are pending. This work was supported by Blueprint Medicines Corporation and F. Hoffmann-La Roche, Ltd, Switzerland (no grant number).
- Published
- 2022
5. Safety and efficacy of pralsetinib in RET fusion-positive non-small-cell lung cancer including as first-line therapy: update from the ARROW trial.
- Author
-
Griesinger F, Curigliano G, Thomas M, Subbiah V, Baik CS, Tan DSW, Lee DH, Misch D, Garralda E, Kim DW, van der Wekken AJ, Gainor JF, Paz-Ares L, Liu SV, Kalemkerian GP, Houvras Y, Bowles DW, Mansfield AS, Lin JJ, Smoljanovic V, Rahman A, Kong S, Zalutskaya A, Louie-Gao M, Boral AL, and Mazières J
- Subjects
- Humans, Proto-Oncogene Proteins c-ret genetics, Pyrazoles therapeutic use, Pyrimidines adverse effects, Adolescent, Adult, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics
- Abstract
Background: RET fusions are present in 1%-2% of non-small-cell lung cancer (NSCLC). Pralsetinib, a highly potent, oral, central nervous system-penetrant, selective RET inhibitor, previously demonstrated clinical activity in patients with RET fusion-positive NSCLC in the phase I/II ARROW study, including among treatment-naive patients. We report an updated analysis from the ARROW study., Patients and Methods: ARROW is a multi-cohort, open-label, phase I/II study. Eligible patients were ≥18 years of age with locally advanced or metastatic solid tumours and an Eastern Cooperative Oncology Group performance status of 0-2 (later 0-1). Patients initiated pralsetinib at the recommended phase II dose of 400 mg once daily until disease progression, intolerance, consent withdrawal, or investigator's decision. The co-primary endpoints (phase II) were overall response rate (ORR) by blinded independent central review and safety., Results: Between 17 March 2017 and 6 November 2020 (data cut-off), 281 patients with RET fusion-positive NSCLC were enrolled. The ORR was 72% [54/75; 95% confidence interval (CI) 60% to 82%] for treatment-naive patients and 59% (80/136; 95% CI 50% to 67%) for patients with prior platinum-based chemotherapy (enrolment cut-off for efficacy analysis: 22 May 2020); median duration of response was not reached for treatment-naive patients and 22.3 months for prior platinum-based chemotherapy patients. Tumour shrinkage was observed in all treatment-naive patients and in 97% of patients with prior platinum-based chemotherapy; median progression-free survival was 13.0 and 16.5 months, respectively. In patients with measurable intracranial metastases, the intracranial response rate was 70% (7/10; 95% CI 35% to 93%); all had received prior systemic treatment. In treatment-naive patients with RET fusion-positive NSCLC who initiated pralsetinib by the data cut-off (n = 116), the most common grade 3-4 treatment-related adverse events (TRAEs) were neutropenia (18%), hypertension (10%), increased blood creatine phosphokinase (9%), and lymphopenia (9%). Overall, 7% (20/281) discontinued due to TRAEs., Conclusions: Pralsetinib treatment produced robust efficacy and was generally well tolerated in treatment-naive patients with advanced RET fusion-positive NSCLC. Results from the confirmatory phase III AcceleRET Lung study (NCT04222972) of pralsetinib versus standard of care in the first-line setting are pending., Competing Interests: Disclosure FG has consulted or provided expert opinion for Amgen, AstraZeneca, Bayer, BMS, Boehringer Ingelheim, Celgene, GSK, Lilly, MSD, Novartis, Pfizer, Roche, Siemens, and Takeda; has received fees from Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, BMS, Celgene, GSK, Lilly, MSD, Novartis, Pfizer, Roche, Siemens, and Takeda; and has received funding for scientific research from Amgen, AstraZeneca, Boehringer Ingelheim, BMS, Celgene, GSK, Lilly, MSD, Novartis, Pfizer, Roche, Siemens, and Takeda. GC has consulted and/or had advisory roles for AstraZeneca, BMS, Boehringer Ingelheim, Daiichi Sankyo, Foundation Medicine, GlaxoSmithKline, Lilly, Novartis, Pfizer, Roche/Genentech, Samsung, and Seattle Genetics; served on speakers’ bureaus for Daiichi Sankyo, Foundation Medicine, Lilly, Novartis, Pfizer, Roche/Genentech, and Samsung; received travel, accommodations, and expenses from Pfizer, Roche/Genentech; received honoraria from Ellipses Pharma and research funding from Merck; and is supported by the OPTIMA [grant number 101034347]. MT has received honoraria for scientific meetings (self) from AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Chugai, Lilly, MSD, Novartis, Pfizer, Roche, and Takeda; advisory-board honoraria (self) from AstraZeneca, BMS, Boehringer Ingelheim, Lilly, MSD, Novartis, Pfizer, Roche, and Takeda; travelling support (self) from AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Chugai, Lilly, MSD, Novartis, Pfizer, Roche, and Takeda; and has received research funding (institution) from AstraZeneca, BMS, Roche, and Takeda. VS reports research funding/grant support for clinical trials from AbbVie, Agensys, Alfa-sigma, Altum, Amgen, Bayer, Berg Health, Biotherapeutics, Blueprint Medicines Corporation, Boston Biomedical, Boston Pharmaceuticals, Celgene, D3, Dragonfly Therapeutics, Exelixis, Fujifilm, GSK, Idera Pharma, Incyte, Inhibrx, Loxo Oncology, MedImmune, MultiVir, Nanocarrier, National Comprehensive Cancer Network, NCI-CTEP, Novartis, Northwest Biotherapeutics, Pfizer, PharmaMar, Roche/Genentech, Takeda, Turning Point Therapeutics, UT MD Anderson Cancer Center, and Vegenics; travel support from ASCO, ESMO, Helsinn, Incyte, Novartis, and PharmaMar; consultancy/advisory board participation for Helsinn, Incyte, Loxo Oncology/Eli Lilly, MedImmune, Novartis, R-Pharma US, QED Pharma; and other relationship with Medscape. CSB has received consulting fees from AstraZeneca, Blueprint Medicines Corporation, Daiichi Sankyo, Turning Point Therapeutics, Guardant, Regeneron, Silverback, and Takeda; and has received research funding to their institution from AbbVie, AstraZeneca, Blueprint Medicines Corporation, Daiichi Sankyo, Genentech Inc., Janssen, Lilly, Loxo Oncology, Novartis, Pfizer, Rain Therapeutics, Spectrum Pharmaceuticals, and Turning Point Therapeutics. DSWT has consulted and/or had advisory roles for AstraZeneca, Bayer, Lilly, Loxo Oncology, Merrimack, Novartis, Pfizer, and Takeda; received honoraria from Boehringer Ingelheim, Merck, and Roche; and research funding to their institution from AstraZeneca, Bayer, GSK, and Novartis. DHL has received personal fees from AbbVie, AstraZeneca, Boehringer Ingelheim, BMS, Chong Keun Dang, CJ Healthcare, Genexine, Janssen, Lilly, Merck, Menarini, MSD, Mundipharma, Novartis, Ono, Pfizer, Roche, Samyang Biopharm, ST Cube, and Takeda; and non-financial support from Blueprint Medicines Corporation and Takeda. DM has consulted and/or had advisory roles at scientific meetings for AstraZeneca, BMS, Boehringer Ingelheim, Lilly, MSD, Novartis, Roche, Sanofi, and Takeda (institution, no personal honoraria). EG has consulted and/or had advisory roles for Alkermes, BMS, Boehringer Ingelheim, Ellipses Pharma, Janssen, NeoMed, Roche, Seattle Genetics, TFS, Thermo Fisher Scientific; served on speakers’ bureaus for MSD, Roche, and Thermo Fisher Scientific; received travel and accommodation expenses from BMS, Glycotope GmbH, Menarini, and MSD; research funding to their institution from Novartis, Roche, and Thermo Fisher Scientific; and is supported by a grant from the ‘la Caixa’ Foundation [grant number LCF/PR/CE07/50610001]. DWK has received travel and accommodation expenses from Amgen and Daiichi Sankyo; and research funding to their institution from Alpha Biopharma, Amgen, AstraZeneca/MedImmune, Boehringer Ingelheim, Daiichi Sankyo, Hanmi, Janssen, Merus, Mirati Therapeutics, MSD, Novartis, Ono Pharmaceutical, Pfizer, Roche/Genentech, Takeda, TP Therapeutics, Xcovery, and Yuhan. AJvdW reports research funding/grant support for clinical trials from AstraZeneca [grant number ESR-16-12212], Boehringer Ingelheim, Pfizer, Roche, and Takeda [grant number 2019N0853/2020N0366]; and consultancy/advisory board participation for AstraZeneca, Boehringer Ingelheim, Janssen, Lilly, Merck, Pfizer, Roche, and Takeda. JFG has an immediate family member who is an employee of Ironwood Pharmaceuticals; has consulted and/or had advisory roles for Agios, Amgen, Array BioPharma, Blueprint Medicines Corporation, BMS, Genentech, Gilead Sciences, Jounce Therapeutics, Lilly, Loxo Oncology, Merck, Mirati, Silverback Therapeutics, GlydeBio, Moderna Therapeutics, Oncorus, Regeneron, Takeda, and Theravance; has stock and ownership in Ironwood Pharmaceuticals; has received honoraria from ARIAD, Incyte, Merck, Novartis, Pfizer, and Takeda; and research funding from Adaptimmune, ALX Oncology, ARIAD, Array BioPharma, AstraZeneca, Blueprint Medicines Corporation, BMS, Genentech, Jounce Therapeutics, Merck, Novartis, and Tesaro. LPA has a leadership role in ALTUM Sequencing and Genomica; served on speakers’ bureaus for AstraZeneca, BMS, Lilly, MSD Oncology, Merck Serono, Pfizer, Roche/Genentech; received travel, accommodation, and expenses from AstraZeneca, BMS, MSD, Pfizer, Roche, and Takeda; honoraria from Amgen, AstraZeneca, Bayer, Blueprint Medicines Corporation, BMS, Celgene, Ipsen, Lilly, Merck Serono, Mirati Therapeutics, MSD, Novartis, Pfizer, PharmaMar, Roche/Genentech, Sanofi, Servier, and Takeda; research funding to their institution from AstraZeneca, BMS, Kura Oncology, MSD, and PharmaMar; other relationships with Roche; and an immediate family member has other relationships with Amgen, Ipsen, Merck Novartis, Pfizer, Sanofi, Servier, and Roche. SVL served as a consultant or advisory board member to Amgen, AstraZeneca, Bayer, BeiGene, Blueprint Medicines Corporation, BMS, Daiichi Sankyo, Eisai, Elevation Oncology, Genentech/Roche, Gilead, Guardant Health, Janssen, Jazz Pharmaceuticals, Lilly, Merck/MSD, Novartis, Regeneron, Sanofi, Takeda, and Turning Point Therapeutics; received research funding (to institution) from Alkermes, Bayer, Blueprint Medicines Corporation, BMS, Elevation Oncology, Genentech, Lilly, Merck, Merus, Pfizer, Rain Therapeutics, RAPT Therapeutics, Turning Point Therapeutics; and is supported by the National Cancer Institute [grant number UM1CA186691]. GPK received research grants from Blueprint Medicines Corporation, Merck, AbbVie, Takeda, Daiichi, and Cullinan. DWB served on an advisory board for Blueprint Medicines Corporation. ASM received research funding from DoD, Mark Foundation, NIH, Novartis, and Verily; honoraria to institution for participation in advisory boards: AbbVie, BeiGene, BMS, Genentech, Inc., Janssen; travel support from: Roche; is a non-remunerated member of the Mesothelioma Applied Research Foundation Board of Directors; and is supported by the Mark Foundation for Cancer Research ASPIRE Award, the National Cancer Institute [grant number R21 CA251923], and Department of Defense Concept Award [grant number W81XWH-22-1-0021]. JJL served as a compensated consultant or advisory board member for Genentech, C4 Therapeutics, Blueprint Medicines Corporation, Nuvalent, Bayer, Elevation Oncology, Novartis, Mirati Therapeutics, and Turning Point Therapeutics; received honorarium and travel support from Pfizer; received institutional research funding from Hengrui Therapeutics, Turning Point Therapeutics, Neon Therapeutics, Relay Therapeutics, Bayer, Elevation Oncology, Roche, Linnaeus Therapeutics, Nuvalent, and Novartis; and received CME funding from OncLive, MedStar Health, and Northwell Health. VSm is an employee and/or equity holder of F. Hoffmann-La Roche, Ltd. AR is an employee and/or equity holder of F. Hoffmann-La Roche, Ltd and equity holder of Merck/MSD. SK is a former employee and/or equity holder of F. Hoffmann-La Roche, Ltd. AZ, MLG and ALB are employees and/or equity holders of Blueprint Medicines Corporation. JM has provided expertise for Amgen, AstraZeneca, Blueprint Medicines Corporation, BMS, Daiichi Sankyo, Hengrui, MSD, Novartis, Pierre Fabre, Roche, and Takeda; and received research funding from AstraZeneca, BMS, Pierre Fabre, and Roche. YH has declared no conflicts of interest. Data sharing The anonymized derived data from this study that underlie the results reported in this article will be made available, beginning 12 months and ending 5 years after this article’s publication, to any investigators who sign a data access agreement and provide a methodologically sound proposal to medinfo@blueprintmedicines.com. The trial protocol will also be made available, as will a data fields dictionary., (Copyright © 2022 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
6. Performance status (PS) as a predictor of poor response to immune checkpoint inhibitors (ICI) in recurrent/metastatic head and neck cancer (RMHNSCC) patients.
- Author
-
Chalker C, Voutsinas JM, Wu QV, Santana-Davila R, Hwang V, Baik CS, Lee S, Barber B, Futran ND, Houlton JJ, Laramore GE, Liao JJ, Parvathaneni U, Martins RG, Eaton KD, and Rodriguez CP
- Subjects
- Humans, Squamous Cell Carcinoma of Head and Neck drug therapy, Immune Checkpoint Inhibitors adverse effects, Retrospective Studies, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Head and Neck Neoplasms drug therapy, Carcinoma drug therapy
- Abstract
Background: Anti-PD1 checkpoint inhibitors (ICI) represent an established standard-of-care for patients with recurrent/metastatic head and neck squamous cell carcinoma (RMHNSCC). Landmark studies excluded patients with ECOG performance status (PS) ≥2; the benefit of ICI in this population is therefore unknown., Methods: We retrospectively reviewed RMHNSCC patients who received 1+ dose of ICI at our institution between 2013 and 2019. Demographic and clinical data were obtained; the latter included objective response (ORR), toxicity, and any unplanned hospitalization (UH). Associations were explored using uni- and multivariate analysis. Overall survival (OS) was estimated using a Cox proportional hazards model; ORR, toxicity, and UH were evaluated with logistic regression., Results: Of the 152 patients, 29 (19%) had an ECOG PS ≥2. Sixty-six (44%) experienced toxicity; 54 (36%) had a UH. A multivariate model for OS containing PS, smoking status, and HPV status demonstrated a strong association between ECOG ≥2 and shorter OS (p < 0.001; HR = 3.30, CI = 2.01-5.41). An association between OS and former (vs. never) smoking was also seen (p < 0.001; HR = 2.17, CI = 1.41-3.35); current smoking did not reach statistical significance. On univariate analysis, poor PS was associated with inferior ORR (p = 0.03; OR = 0.25, CI = 0.06-0.77) and increased UH (p = 0.04; OR = 2.43, CI = 1.05-5.71). There was no significant association between toxicity and any patient characteristic., Conclusions: We observed inferior OS, ORR, and rates of UH among ICI-treated RMHNSCC patients with ECOG 2/3. Our findings help frame discussion of therapeutic options in this poor-risk population., (© 2022 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
7. New Immuno-oncology Targets and Resistance Mechanisms.
- Author
-
Tokaz MC, Baik CS, Houghton AM, and Tseng D
- Subjects
- Humans, Immune Checkpoint Inhibitors, Immunotherapy, Medical Oncology, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung etiology, Lung Neoplasms pathology
- Abstract
Opinion Statement: Immune checkpoint inhibition (ICI) has revolutionized the field of non-small cell lung cancer (NSCLC); currently, most patients with advanced disease receive upfront ICI either alone or in combination with chemotherapy. These advances have recently extended into early-stage NSCLC, with ICI incorporation into neoadjuvant and adjuvant treatment regimens. However, despite these successes, immunotherapy (IO) resistance remains a fundamental challenge in NSCLC, introducing a central quandary of how to precisely select the appropriate IO therapy or IO combination therapy for each individual patient. To address this vital need in the field, there has been an explosion of research in immuno-oncology to identify mechanisms of resistance, ranging from genomic alterations in the tumor to immunosuppressive conditions in the tumor microenvironment (TME). There remain many questions about how this complex interplay between the tumor and the immune microenvironment translates into clinical phenotypes of primary and acquired resistance. In NSCLC, a number of novel therapeutics are being developed to prevent and overcome resistance to ICI. Particular promise has been shown with therapeutics targeting novel T cell immune checkpoint inhibitors and targeting innate immune cells in the TME, chief among these cells are natural killer cells, neutrophils, and macrophages. Further research into tissue-based and non-invasive biomarkers that can be prospectively integrated into therapeutic trial design will be critical to advance the field's understanding of individual resistance patterns and enable the ultimate goal of precision immuno-oncology., (© 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
8. HER2 in Non-Small Cell Lung Cancer: A Review of Emerging Therapies.
- Author
-
Uy NF, Merkhofer CM, and Baik CS
- Abstract
Human epidermal growth factor receptor 2 (HER2), a member of the ERBB family of tyrosine kinase receptors, has emerged as a therapeutic target of interest for non-small cell lung cancer (NSCLC) in recent years. Activating HER2 alterations in NSCLC include gene mutations, gene amplifications, and protein overexpression. In particular, the HER2 exon 20 mutation is now a well clinically validated biomarker. Currently, there are limited targeted therapies approved for NSCLC patients with HER2 alterations. This remains an unmet clinical need, as HER2 alterations are present in 7-27% of de novo NSCLC and may serve as a resistance mechanism in up to 10% of EGFR mutated NSCLC. There has been an influx of research on antibody-drug conjugates (ADCs), monoclonal antibodies, and tyrosine kinase inhibitors (TKIs) with mixed results. The most promising therapies are ADCs (trastuzumab-deruxtecan) and novel TKIs targeting exon 20 mutations (poziotinib, mobocertinib and pyrotinib); both have resulted in meaningful anti-tumor efficacy in HER2 mutated NSCLC. Future studies on HER2 targeted therapy will need to define the specific HER2 alteration to better select patients who will benefit, particularly for HER2 amplification and overexpression. Given the variety of HER2 targeted drugs, sequencing of these agents and optimizing combination therapies will depend on more mature efficacy data from clinical trials and toxicity profiles. This review highlights the challenges of diagnosing HER2 alterations, summarizes recent progress in novel HER2-targeted agents, and projects next steps in advancing treatment for the thousands of patients with HER2 altered NSCLC.
- Published
- 2022
- Full Text
- View/download PDF
9. Clinical Outcomes With Dabrafenib Plus Trametinib in a Clinical Trial Versus Real-World Standard of Care in Patients With BRAF -Mutated Advanced NSCLC.
- Author
-
Johnson BE, Baik CS, Mazieres J, Groen HJM, Melosky B, Wolf J, Zadeh Vosta Kolaei FA, Wu WH, Knoll S, Ktiouet Dawson M, Johns A, and Planchard D
- Abstract
Introduction: BRAF mutations are rare in patients with NSCLC, and treatment options are limited. Dabrafenib plus trametinib (dab-tram) was approved for BRAF
V600 -mutated advanced NSCLC (aNSCLC), based on results from a phase 2 study (NCT01336634). This retrospective analysis compared the effectiveness of dab-tram, based on previously reported clinical trial data, versus real-world standard of care in patients with BRAF -mutated aNSCLC., Methods: Real-world cohorts were derived from a deidentified real-world database (2011-2020) and included patients with BRAF -mutated aNSCLC receiving first-line platinum-based chemotherapy (PBC), first-line immune checkpoint inhibitors (ICIs) plus PBC, or second-line ICIs. Weighting by odds was used to estimate the average treatment effect of the treated., Results: For first-line dab-tram versus PBC, the hazard ratio (HR; 95% confidence interval) for death in unweighted and weighted analyses was 0.65 (0.39-1.1) and 0.51 (0.29-0.92; p = 0.03), respectively; unweighted and weighted median overall survival was 17.3 (12.3-40.2) versus 14.5 (9.2-19.6) months and 17.3 (14.6-not reached) versus 9.7 (6.4-19.6) months, respectively. Hazard ratio of death in unweighted and weighted analyses was 0.56 (0.29-1.1) and 0.57 (0.28-1.17), respectively, with first-line dab-tram versus PBC plus ICI, and 0.65 (0.39-1.07) and not reported (Cox proportional-hazards assumption violated), respectively, with second-line dab-tram versus ICI., Conclusions: In this indirect comparison in patients with BRAF -mutated aNSCLC, the risk of death was lower and median overall survival was longer with first-line dab-tram versus PBC. In analyses of dab-tram versus first-line PBC plus ICI or second-line ICI, sample sizes were small and findings were inconclusive with overlapping confidence intervals. Despite some limitations, the study provides useful data for this rare patient population., (© 2022 The Authors.)- Published
- 2022
- Full Text
- View/download PDF
10. High End-of-Life Health Care Utilization in a Contemporary Cohort of Head and Neck Cancer Patients Treated with Immune Checkpoint Inhibitors.
- Author
-
Chalker C, Santana-Davila R, Voutsinas JM, Wu QV, Hwang V, Baik CS, Lee S, Barber B, Futran ND, Houlton JJ, Laramore GE, Liao JJ, Parvathaneni U, Martins RG, Eaton KD, and Rodriguez CP
- Subjects
- Death, Humans, Patient Acceptance of Health Care, Retrospective Studies, Head and Neck Neoplasms drug therapy, Immune Checkpoint Inhibitors
- Abstract
Background/Objective: End-of-life health care utilization (EOLHCU) is largely uncharacterized among patients with recurrent/metastatic head and neck squamous cell carcinomas (RMHNSCC), particularly now that immune checkpoint inhibitors (ICI) have been introduced to the treatment landscape. We examined this in a single-institution, retrospective study. Design/Settings: We utilized a database of deceased, ICI-treated RMHNSCC patients to obtain demographic and EOLHCU data, the latter of which included advanced care plan documentation (ACPD) and systemic therapy or emergency room (ER)/hospital/intensive care unit (ICU) admission within 30 days of death (DOD). This was compared with a cohort of deceased thoracic malignancy (TM) patients in an exploratory analysis. Multivariate analysis was performed to examine for association between patient factors (such as age, Eastern Cooperative Oncology Group (ECOG) performance status, or smoking status) and overall survival (OS); associations between the said patient factors and EOLHCU were also evaluated. This study was conducted at an academic, tertiary center in the United States. Results: The RMHNSCC patients ( n = 74) were more likely to have ACPD ( p < 0.01), an emergency department visit ( p < 0.01), and/or hospital admission ( p < 0.01) within 30 DOD relative to the TM group. There was no difference in ICU admissions, ICU deaths, or systemic therapy at end of life (EOL). The OS declined in association with ECOG performance status (PS) and smoking. No association was observed between patient factors and any EOLHCU metric. Conclusions: At our center, patients with ICI-treated RMHNSCC have higher rates of both ACPD and EOLHCU, suggesting high symptom burden and representing opportunities for further study into supportive care augmentation.
- Published
- 2022
- Full Text
- View/download PDF
11. Prognostic Value of Early Fluorodeoxyglucose-Positron Emission Tomography Response Imaging and Peripheral Immunologic Biomarkers: Substudy of a Phase II Trial of Risk-Adaptive Chemoradiation for Unresectable Non-Small Cell Lung Cancer.
- Author
-
Bowen SR, Hippe DS, Thomas HM, Sasidharan B, Lampe PD, Baik CS, Eaton KD, Lee S, Martins RG, Santana-Davila R, Chen DL, Kinahan PE, Miyaoka RS, Vesselle HJ, Houghton AM, Rengan R, and Zeng J
- Abstract
Purpose: We sought to examine the prognostic value of fluorodeoxyglucose-positron emission tomography (PET) imaging during chemoradiation for unresectable non-small cell lung cancer for survival and hypothesized that tumor PET response is correlated with peripheral T-cell function., Methods and Materials: Forty-five patients with American Joint Committee on Cancer version 7 stage IIB-IIIB non-small cell lung cancer enrolled in a phase II trial and received platinum-doublet chemotherapy concurrent with 6 weeks of radiation (NCT02773238). Fluorodeoxyglucose-PET was performed before treatment start and after 24 Gy of radiation (week 3). PET response status was prospectively defined by multifactorial radiologic interpretation. PET responders received 60 Gy in 30 fractions, while nonresponders received concomitant boosts to 74 Gy in 30 fractions. Peripheral blood was drawn synchronously with PET imaging, from which germline DNA sequencing, T-cell receptor sequencing, and plasma cytokine analysis were performed., Results: Median follow-up was 18.8 months, 1-year overall survival (OS) 82%, 1-year progression-free survival 53%, and 1-year locoregional control 88%. Higher midtreatment PET total lesion glycolysis was detrimental to OS (1 year 87% vs 63%, P < .001), progression-free survival (1 year 60% vs 26%, P = .044), and locoregional control (1 year 94% vs 65%, P = .012), even after adjustment for clinical/treatment factors. Twenty-nine of 45 patients (64%) were classified as PET responders based on a priori definition. Higher tumor programmed death-ligand 1 expression was correlated with response on PET ( P = .017). Higher T-cell receptor richness and clone distribution slope were associated with improved OS ( P = .018-0.035); clone distribution slope was correlated with PET response ( P = .031)., Conclusions: Midchemoradiation PET imaging is prognostic for survival; PET response may be linked to tumor and peripheral T-cell biomarkers., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
12. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study.
- Author
-
Subbiah V, Hu MI, Wirth LJ, Schuler M, Mansfield AS, Curigliano G, Brose MS, Zhu VW, Leboulleux S, Bowles DW, Baik CS, Adkins D, Keam B, Matos I, Garralda E, Gainor JF, Lopes G, Lin CC, Godbert Y, Sarker D, Miller SG, Clifford C, Zhang H, Turner CD, and Taylor MH
- Subjects
- Aged, Carcinoma, Neuroendocrine genetics, Carcinoma, Neuroendocrine pathology, Female, Humans, Male, Middle Aged, Neoplasm Metastasis, Prognosis, Survival Rate, Thyroid Neoplasms genetics, Thyroid Neoplasms pathology, Antineoplastic Agents therapeutic use, Carcinoma, Neuroendocrine drug therapy, Mutation, Proto-Oncogene Proteins c-ret genetics, Pyrazoles therapeutic use, Pyridines therapeutic use, Pyrimidines therapeutic use, Thyroid Neoplasms drug therapy
- Abstract
Background: Oncogenic alterations in RET represent important therapeutic targets in thyroid cancer. We aimed to assess the safety and antitumour activity of pralsetinib, a highly potent, selective RET inhibitor, in patients with RET-altered thyroid cancers., Methods: ARROW, a phase 1/2, open-label study done in 13 countries across 71 sites in community and hospital settings, enrolled patients 18 years or older with RET-altered locally advanced or metastatic solid tumours, including RET-mutant medullary thyroid and RET fusion-positive thyroid cancers, and an Eastern Co-operative Oncology Group performance status of 0-2 (later limited to 0-1 in a protocol amendment). Phase 2 primary endpoints assessed for patients who received 400 mg once-daily oral pralsetinib until disease progression, intolerance, withdrawal of consent, or investigator decision, were overall response rate (Response Evaluation Criteria in Solid Tumours version 1.1; masked independent central review) and safety. Tumour response was assessed for patients with RET-mutant medullary thyroid cancer who had received previous cabozantinib or vandetanib, or both, or were ineligible for standard therapy and patients with previously treated RET fusion-positive thyroid cancer; safety was assessed for all patients with RET-altered thyroid cancer. This ongoing study is registered with clinicaltrials.gov, NCT03037385, and enrolment of patients with RET fusion-positive thyroid cancer was ongoing at the time of this interim analysis., Findings: Between Mar 17, 2017, and May 22, 2020, 122 patients with RET-mutant medullary and 20 with RET fusion-positive thyroid cancers were enrolled. Among patients with baseline measurable disease who received pralsetinib by July 11, 2019 (enrolment cutoff for efficacy analysis), overall response rates were 15 (71%) of 21 (95% CI 48-89) in patients with treatment-naive RET-mutant medullary thyroid cancer and 33 (60%) of 55 (95% CI 46-73) in patients who had previously received cabozantinib or vandetanib, or both, and eight (89%) of nine (95% CI 52-100) in patients with RET fusion-positive thyroid cancer (all responses confirmed for each group). Common (≥10%) grade 3 and above treatment-related adverse events among patients with RET-altered thyroid cancer enrolled by May 22, 2020, were hypertension (24 patients [17%] of 142), neutropenia (19 [13%]), lymphopenia (17 [12%]), and anaemia (14 [10%]). Serious treatment-related adverse events were reported in 21 patients (15%), the most frequent (≥2%) of which was pneumonitis (five patients [4%]). Five patients [4%] discontinued owing to treatment-related events. One (1%) patient died owing to a treatment-related adverse event., Interpretation: Pralsetinib is a new, well-tolerated, potent once-daily oral treatment option for patients with RET-altered thyroid cancer., Funding: Blueprint Medicines., Competing Interests: Declaration of interests VS reports grants from Blueprint Medicines and LOXO Oncology–Eli Lilly; research funding or grant support from Roche–Genentech, Novartis, Bayer, GlaxoSmithKline (GSK), Nanocarrier, Vegenics, Northwest Biotherapeutics, Berg Heath, Incyte, Fujifilm, PharmaMar, D3, Pfizer, MultiVir, Amgen, AbbVie, Alfasigma, Agensys, Boston Biomedical, Idera Pharma, Inhibrx, Exelixis, Medimmune, Altum, Dragonfly Therapeutics, Takeda, Immunogen, National Comprehensive Cancer Network (NCCN), NCI-CTEP and UT MD Anderson Cancer Center, Turning Point Therapeutics, and Boston Pharmaceuticals, travel support from Novartis, PharmaMar, ASCO, ESMO, Helsinn, and Incyte, and consultancy or advisory board participation for Helsinn, LOXO Oncology–Eli Lilly, R-Pharma US, Incyte, QED Pharma, Medimmune, and Novartis, and another relationship with Medscape. MIH reports advisory board roles for Eli Lilly, LOXO Oncology, and Blueprint Medicines, and research funding from Eli Lilly. LJW reports personal fees and other support from Blueprint Medicines; personal fees from Bayer, Blueprint Medicines, Exelixis, Genentech, Eli Lilly, and Merck, and non-financial support from Eli Lilly and Merck. MS reports grants from AstraZeneca and Bristol-Myers Squibb (BMS), and personal fees from AstraZeneca, Boehringer Ingelheim, BMS, GSK, Janssen, MorphoSys, Novartis, Roche, Takeda, Amgen, and MSD. ASM reports consulting or advisory board roles (with honoraria to his institution) for Janssen, Genentech, BMS, AbbVie, AstraZeneca, travel or accommodation or expenses from AbbVie and Roche, and research funding from Novartis, NIH, Mark Foundation, and is a non-remunerated board member of the Mesothelioma Applied Research Foundation. GC reports personal fees from Roche, Pfizer, Novartis, Eli Lilly, Foundation Medicine, BMS, Samsung, AstraZeneca, Daiichi Sankyo, Boehringer Ingelheim, GSK, and Seagen, non-financial support from Roche and Pfizer, grants from Merck, and other support from Ellipses Pharma. MSB has nothing to disclose. VWZ reports personal fees from AstraZeneca, Blueprint Medicines, Roche-Foundation Medicine, Roche–Genentech, Takeda, Turning Point Therapeutics, and Xcovery. SL reports personal fees from Eisai and Eli Lilly. DWB reports personal fees from Blueprint Medicines. CSB reports grants from Blueprint Medicines, Daiichi Sankyo, Celgene, Pfizer, Lilly Oncology, AbbVie, Rain Therapeutics, Spectrum Pharmaceuticals, and personal fees from AstraZeneca, Turning Point Therapeutics, NCCN, and Takeda. DA reports grants and personal fees from Blueprint Medicines; grants from Exelixis, Kura Oncology, Merck, Cue, Aduro, Eli Lilly, Pfizer, Celgene–BMS, Novartis, AstraZeneca, Atara Bio, Cellex, Innate, Sensei, Matrix Biomed, Hookipa, CoFactor, Medimmune, and ISA, and personal fees from Exelixis, Merck, Cue, twoXAR, Vaccinex, Zilio, and TargImmune. BK has nothing to disclose. IM reports grants from an ESMO Research Fellowship sponsored by Roche, and personal fees for speaker bureau participation from MSD. EG reports personal fees from Genentech, Hoffman–LaRoche, Ellipses Pharma, Neomed Therapeutics, Boehringer Ingelheim, Janssen, Seattle Genetics, TFS, Alkermes, ThermoFisher, BMS, and MSD, grants from Menarini and Glycotope, and other support from Novartis, Roche, and ThermoFisher. JFG reports personal fees from BMS, Roche–Genentech, Merck, Takeda, LOXO Oncology–Lilly, Blueprint Medicines, Oncorus, Regeneron, Gilead, AstraZeneca, Pfizer, Novartis, and Moderna, grants from Takeda and Novartis, and institutional research support from BMS, LOXO Oncology–Lilly, Blueprint Medicines, Novartis, Moderna, Alexo, Tesaro, Jounce, Adaptimmune Therapeutics, and an immediate family member who is an employee of and holds equity in Ironwood Pharmaceuticals. GL reports other support from Bayer, Blueprint Medicines, Cue Biopharma, Eisai, Exelixis, Genentech, Eli Lilly, LOXO Oncology, Merck, and Rakuten Medical. C-CL reports personal fees from BeiGene, Blueprint Medicines, Boehringer Ingelheim, BMS, Daiichi Sankyo, Eli Lilly, Novartis, Takeda, and Roche. YG has nothing to disclose. DS reports grants from Blueprint Medicines, personal fees from MSD, Eisai, Ipsen, Bayer, AstraZeneca, Surface Oncology, and non-financial support from Eisai, Ipsen, and Monia Therapeutics. SGM, CC, and HZ are employees of and hold equity interest in Blueprint Medicines. CDT is a former employee of and holds equity interest in Blueprint Medicines. MHT reports other support from Bayer, Blueprint Medicines, Novartis, Sanofi–Genzyme, ArQule, BMS, Eisai, Merck, Array BioPharma, LOXO Oncology, Abreos Biosciences, and Arch Oncology., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): a multi-cohort, open-label, phase 1/2 study.
- Author
-
Gainor JF, Curigliano G, Kim DW, Lee DH, Besse B, Baik CS, Doebele RC, Cassier PA, Lopes G, Tan DSW, Garralda E, Paz-Ares LG, Cho BC, Gadgeel SM, Thomas M, Liu SV, Taylor MH, Mansfield AS, Zhu VW, Clifford C, Zhang H, Palmer M, Green J, Turner CD, and Subbiah V
- Subjects
- Aged, Antineoplastic Agents adverse effects, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Male, Middle Aged, Protein Kinase Inhibitors adverse effects, Pyrazoles adverse effects, Pyridines adverse effects, Pyrimidines adverse effects, Time Factors, Treatment Outcome, Antineoplastic Agents therapeutic use, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Gene Fusion, Lung Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-ret antagonists & inhibitors, Proto-Oncogene Proteins c-ret genetics, Pyrazoles therapeutic use, Pyridines therapeutic use, Pyrimidines therapeutic use
- Abstract
Background: Oncogenic alterations in RET have been identified in multiple tumour types, including 1-2% of non-small-cell lung cancers (NSCLCs). We aimed to assess the safety, tolerability, and antitumour activity of pralsetinib, a highly potent, oral, selective RET inhibitor, in patients with RET fusion-positive NSCLC., Methods: ARROW is a multi-cohort, open-label, phase 1/2 study done at 71 sites (community and academic cancer centres) in 13 countries (Belgium, China, France, Germany, Hong Kong, Italy, Netherlands, Singapore, South Korea, Spain, Taiwan, the UK, and the USA). Patients aged 18 years or older with locally advanced or metastatic solid tumours, including RET fusion-positive NSCLC, and an Eastern Cooperative Oncology Group performance status of 0-2 (later limited to 0-1 in a protocol amendment) were enrolled. In phase 2, patients received 400 mg once-daily oral pralsetinib, and could continue treatment until disease progression, intolerance, withdrawal of consent, or investigator decision. Phase 2 primary endpoints were overall response rate (according to Response Evaluation Criteria in Solid Tumours version 1·1 and assessed by blinded independent central review) and safety. Tumour response was assessed in patients with RET fusion-positive NSCLC and centrally adjudicated baseline measurable disease who had received platinum-based chemotherapy or were treatment-naive because they were ineligible for standard therapy. This ongoing study is registered with ClinicalTrials.gov, NCT03037385, and enrolment of patients with treatment-naive RET fusion-positive NSCLC was ongoing at the time of this interim analysis., Findings: Of 233 patients with RET fusion-positive NSCLC enrolled between March 17, 2017, and May 22, 2020 (data cutoff), 92 with previous platinum-based chemotherapy and 29 who were treatment-naive received pralsetinib before July 11, 2019 (efficacy enrolment cutoff); 87 previously treated patients and 27 treatment-naive patients had centrally adjudicated baseline measurable disease. Overall responses were recorded in 53 (61%; 95% CI 50-71) of 87 patients with previous platinum-based chemotherapy, including five (6%) patients with a complete response; and 19 (70%; 50-86) of 27 treatment-naive patients, including three (11%) with a complete response. In 233 patients with RET fusion-positive NSCLC, common grade 3 or worse treatment-related adverse events were neutropenia (43 patients [18%]), hypertension (26 [11%]), and anaemia (24 [10%]); there were no treatment-related deaths in this population., Interpretation: Pralsetinib is a new, well-tolerated, promising, once-daily oral treatment option for patients with RET fusion-positive NSCLC., Funding: Blueprint Medicines., Competing Interests: Declaration of interests JFG reports personal fees from Bristol-Myers Squibb (BMS), Roche–Genentech, Merck, Takeda, LOXO Oncology–Lilly, Blueprint Medicines, Oncorus, Regeneron, Gilead, AstraZeneca, Pfizer, Novartis, and Moderna, grants from Takeda and Novartis, and institutional research support from BMS, LOXO Oncology–Lilly, Blueprint Medicines, Novartis, Moderna, Alexo, Tesaro, Jounce, Adaptimmune Therapeutics, and an immediate family member who is an employee of and holds equity in Ironwood Pharmaceuticals, all outside the submitted work. GC reports personal fees from Roche, Pfizer, Novartis, Eli Lilly, Foundation Medicine, BMS, Samsung, AstraZeneca, Daiichi-Sankyo, Boehringer Ingelheim, GlaxoSmithKline (GSK), and Seagen, non-financial support from Roche and Pfizer, grants from Merck, and other support from Ellipsis, all outside the submitted work. D-WK reports grants and non-financial support from Blueprint Medicines during the conduct of the study; grants from Alpha Biopharma, Amgen, AstraZeneca–Medimmune, Boehringer Ingelheim, Daiichi-Sankyo, Hanmi, Janssen, Meurs, Mirati Therapeutics, MSD, Novartis, Ono Pharmaceutical, Pfizer, Roche–Genentech, Takeda, Turning Point Therapeutics, Xcovery, and Yuhan, and non-financial support from Amgen and Daiichi-Sankyo, all outside the submitted work. DHL reports personal fees from AstraZeneca, Boehringer Ingelheim, BMS, CJ Healthcare, Eli Lilly, ChongKeunDang, Janssen, Merck, MSD, Mundipharma, Novartis, Ono Pharmaceutical, Pfizer, Roche, Samyang Biopharm, ST Cube, AbbVie, Takeda, Genexine, Menarini, and BC Pharma, and non-financial support from Takeda and Blueprint Medicines, all outside the submitted work. BB reports grants from AbbVie, Amgen, AstraZeneca, BeiGene, Blueprint Medicines, BMS, Boehringer Ingelheim, Celgene, Cristal Therapeutics, Daiichi-Sankyo, Eli Lilly, GSK, Inivata, Janssen, Onxeo, OSE Immunotherapeutics, Pfizer, Roche–Genentech, Sanofi, Takeda, Tolero Pharmaceuticals, 4D Pharma, Aptitude Health, and Cergentis. CSB reports grants from Blueprint Medicines, Daiichi-Sankyo, Celgene, Pfizer, Lilly Oncology, AbbVie, Rain Therapeutics, and Spectrum Pharmaceuticals, and personal fees from AstraZeneca, Turning Point Therapeutics, National Comprehensive Cancer Network, and Takeda, all outside the submitted work. RCD reports consulting–advisory roles for Blueprint Medicines, Rain Therapeutics, Roche–Genentech, Green Peptide, Anchiano, Takeda, AstraZeneca, and Bayer, and travel support from Blueprint Medicines, Rain Therapeutics, Roche–Genentech, Green Peptide, and Takeda, holds stock or has other ownership in Rain Therapeutics, and has patents–royalties from Rain Therapeutics and Abbott Molecular, all outside the submitted work. PAC reports other support from Blueprint Medicines during the conduct of the study; personal fees from Blueprint Medicines, Amgen, Merk Serono, Novartis, and Roche–Genentech, non-financial support from MSD, Novartis, AstraZeneca, and Plexxikon, grants from MSD and Novartis, and other support from AbbVie, Amgen, Bayer, BMS, Merck Serono, MSD, Novartis, Roche–Genentech, GSK, Janssen, Eli Lilly, Netris Pharma, AstraZeneca, Celgene, Taiho, Toray, LOXO Oncology, and Innate Pharma, all outside the submitted work. GL reports consultant roles for Bayer, Blueprint Medicines, Cue Biopharma, Eisai, Exelixis, Roche–Genentech, Eli Lilly, LOXO Oncology, Merck, and Rakuten Medical, all outside the submitted work. DSWT reports personal fees from Novartis, Bayer, Boehringer Ingelheim, AstraZeneca, Eli Lilly, LOXO Oncology, Merck, Pfizer, Roche, Takeda, and Merrimack, and grants from Novartis, Bayer, AstraZeneca, Pfizer, and GSK, all outside the submitted work. EG reports personal fees from Roche–Genentech, Hoffman–La Roche, Ellipses Pharma, Neomed Therapeutics, Boehringer Ingelheim, Janssen, Seattle Genetics, TFS, Alkermes, Thermo Fisher, BMS, and MSD, grants from Menarini and Glycotope, and other support from Novartis, Roche, and Thermo Fisher, all outside the submitted work. LGP-A reports personal fees from Eli Lilly, MSD, Roche, PharmaMar, Merck, AstraZeneca, Novartis, Servier, Amgen, Pfizer, Ipsen, Sanofi, Bayer, Blueprint Medicines, BMS, and Mirati, grants from MSD, AstraZeneca, Pfizer, and BMS, is a board member of Genomica, and is a board member and cofounder of Altum Sequencing, all outside the submitted work. BCC reports grants from Novartis, Bayer, AstraZeneca, MOGAM Institute, Dong-A ST, Champions Oncology, Janssen, Yuhan, Ono Pharmaceuticals, Dizal Pharma, MSD, AbbVie, MedPacto, GI Innovation, Eli Lilly, Blueprint Medicines, and Interpark Bio Convergence Center, personal fees from Novartis, AstraZeneca, Boehringer Ingelheim, Roche, BMS, Ono Pharmaceuticals, Yuhan, Pfizer, Eli Lilly, Janssen, Takeda, MSD, MedPacto, Blueprint Medicines, TheraCanVac, Gencurix, Bridgebio Therapeutics, KANAPH Therapeutics, Cyrus Therapeutics, Interpark Bio Convergence Center, Guardant Health, and Oscotec, other support from Champions Oncology, is a board member of Gencurix and Interpark Bio Convergence Center, and is a founder of DAAN Biotherapeutics, all outside the submitted work. SMG reports personal fees from Blueprint Medicines, Roche–Genentech, AstraZeneca, Janssen, Merck, BMS, Pfizer, Eli Lilly, Novartis, and Daiichi-Sankyo, grants from Merck, and is a compensated member of the independent data monitoring committee for a phase 3 trial sponsored by AstraZeneca, all outside the submitted work. MT reports grants from Blueprint Medicines during the conduct of the submitted work; personal fees and non-financial support from AbbVie, AstraZeneca, BMS, Boehringer Ingelheim, Celgene, Chugai, Eli Lilly, Novartis, Pfizer, Roche, and Takeda, and grants from AstraZeneca, BMS, and Takeda, all outside the submitted work. SVL reports grants from Alkermes, Bayer, Blueprint Medicines, BMS, Roche–Genentech, Eli Lilly, Lycera, Merck/MSD, Molecular Partners, Pfizer, Rain Therapeutics, RAPT, Turning Point Therapeutics, Corvus, Debiopharm, Merus, and Elevation Oncology, and personal fees from AstraZeneca, Blueprint Medicines, BMS, G1 Therapeutics, Roche–Genentech, Guardant Health, Inivata, Janssen, Jazz, Eli Lilly, Merck–MSD, PharmaMar, Pfizer, Regeneron, Takeda, BeiGene, Catalyst, Amgen, and Daiichi-Sankyo, all outside the submitted work. MHT reports consulting or advisory roles for Bayer, Blueprint Medicines, Novartis, Sanofi–Genzyme, ArQule, BMS, Eisai, Merck, Array BioPharma, and LOXO Oncology, travel support from Bayer, Blueprint Medicines, Novartis, Sanofi–Genzyme, ArQule, BMS, Eisai, Array BioPharma, and LOXO Oncology, speaker bureau roles for BMS, Eisai, and Merck, and institutional research funding from Abreos Biosciences and Arch Oncology, all outside the submitted work. ASM reports honoraria to his institution for consulting or advisory roles from Janssen, Roche–Genentech, BMS, AbbVie, AstraZeneca, travel, accommodation, and expenses from AbbVie and Roche, research funding from Novartis, NIH, Verily, and Mark Foundation, and is a non-remunerated board member for the Mesothelioma Applied Research Foundation. VWZ reports personal fees from AstraZeneca, Blueprint Medicines, Roche-Foundation Medicine, Roche–Genentech, Takeda, Turning Point Therapeutics, and Xcovery, all outside the submitted work. CC and HZ are employees of and hold equity interest in Blueprint Medicines. MP is a former employee of and holds equity interest in Blueprint Medicines and reports other support from C4 Therapeutics outside the submitted work. JG reports consulting or advisory roles for Blueprint Medicines, Tesaro, Genocea, and Arvinas. CDT is a former employee of and holds equity interest in Blueprint Medicines. VS reports grants from Blueprint Medicines and LOXO Oncology–Eli Lilly during the conduct of the study; research funding or grant support for clinical trials from Roche–Genentech, Novartis, Bayer, GSK, Nanocarrier, Vegenics, Northwest Biotherapeutics, Berghealth, Incyte, Fujifilm, PharmaMar, D3, Pfizer, Multivir, Amgen, AbbVie, Alfasiga, Agensys, Boston Biomedical, Idera Pharma, Inhibrx, Exelixis, MedImmune, Altum, Dragonfly Therapeutics, Takeda, Immunogen, National Comprehensive Cancer Network, NCI-CTEP, UT MD Anderson Cancer Center, Turning Point Therapeutics, and Boston Pharmaceuticals, travel support from Novartis, PharmaMar, ASCO, ESMO, Helsinn, and Incyte, consultancy or advisory board participation for Helsinn, LOXO Oncology–Eli Lilly, R-Pharma US, Incyte, QED Pharma, MedImmune, and Novartis, and other support from Medscape. All other authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
14. Durable Response of Leptomeningeal Disease With Osimertinib and Pemetrexed in EGFR -Mutated Metastatic NSCLC: A Case Report.
- Author
-
Merkhofer CM and Baik CS
- Abstract
Optimal management of EGFR -mutated NSCLC with leptomeningeal (LM) disease progression through EGFR tyrosine kinase inhibitor remains unclear. We present a 39-year-old man with EGFR -mutated NSCLC and LM disease progression through osimertinib 80 mg daily, with subsequent durable radiographic and symptomatic response to systemic pemetrexed in combination with osimertinib. This builds on the limited data evaluating LM disease response to systemic pemetrexed and lends further support to consideration of this treatment strategy., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
15. Duration of Targeted Therapy in Patients With Advanced Non-small-cell Lung Cancer Identified by Circulating Tumor DNA Analysis.
- Author
-
Reckamp KL, Patil T, Kirtane K, Rich TA, Espenschied CR, Weipert CM, Raymond VM, Santana-Davila R, Doebele RC, and Baik CS
- Subjects
- Adenocarcinoma of Lung drug therapy, Adenocarcinoma of Lung genetics, Adenocarcinoma of Lung pathology, Adult, Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Squamous Cell drug therapy, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell pathology, Circulating Tumor DNA analysis, Female, Follow-Up Studies, Humans, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Male, Middle Aged, Prognosis, Retrospective Studies, Survival Rate, Time Factors, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor antagonists & inhibitors, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung pathology, Circulating Tumor DNA genetics, Lung Neoplasms pathology, Molecular Targeted Therapy
- Abstract
Background: Outcomes of therapy targeting molecular driver alterations detected in advanced non-small-cell lung (NSCLC) using circulating tumor DNA (ctDNA) have not been widely reported in patients who are targeted therapy-naive., Patients and Methods: We performed a multicenter retrospective review of patients with unresectable stage IIIB to IV NSCLC who received matched therapy after a targetable driver alteration was identified using a commercial ctDNA assay through usual clinical care. Eligible patients must not have received targeted therapy prior to ctDNA testing (prior chemotherapy or immunotherapy was permitted). Kaplan-Meier analysis was used to estimate the median duration of targeted therapy. Patients still on targeted therapy were censored at last follow-up., Results: Seventy-six patients met inclusion criteria. The median age of diagnosis of NSCLC was 64.5 years (range, 31-87 years), 67% were female, 74% were never-smokers, and 97% had adenocarcinoma histology. Twenty-one (28%) patients received systemic treatment prior to targeted therapy, including chemotherapy (n = 17), immunotherapy (n = 5), and/or a biologic (n = 4). Thirty-three (43%) patients remain on targeted therapy at the time of data analysis. The median time on targeted therapy was similar to what has been reported for tissue-detected oncogenic driver mutations in the targeted therapy-naive setting., Conclusions: Patients with ctDNA-detected drivers had durable time on targeted therapy. These treatment outcomes data compliment previous studies that have shown enhanced targetable biomarker discovery rates and high tissue concordance of ctDNA testing when incorporated at initial diagnosis of NSCLC. Identification of NSCLC driver mutations using well-validated ctDNA assays can be used for clinical decision-making and targeted therapy assignment., (Copyright © 2020. Published by Elsevier Inc.)
- Published
- 2020
- Full Text
- View/download PDF
16. Outcomes Among Homeless Patients With Non-Small-Cell Lung Cancer: A County Hospital Experience.
- Author
-
Concannon KF, Thayer JH, Wu QV, Jenkins IC, Baik CS, and Linden HM
- Subjects
- Hospitals, County, Humans, Retrospective Studies, United States, Washington epidemiology, Carcinoma, Non-Small-Cell Lung, Lung Neoplasms
- Abstract
Purpose: Lung cancer remains the leading cause of cancer death in the United States, with outcomes likely worsened by the presence of poorer outcomes among vulnerable populations such as the homeless. We hypothesized that homeless patients experience delays in biopsy, decreased appointment adherence, and increased overall mortality rates., Methods: We conducted a retrospective electronic medical record-based review of all patients with non-small-cell lung cancer (NSCLC; N = 133) between September 2012 and September 2018 at an academic county hospital in Seattle, Washington., Results: Of the 133 patients treated for NSCLC, 22 (17%) were homeless at the time of their treatment. Among homeless patients with localized lung cancer, the mean time from radiographic finding to biopsy was 248 days, compared with 116 days among housed patients ( P = .37). Homeless patients with advanced disease missed a mean of 26% of appointments in the year after diagnosis, compared with 16% among housed patients ( P = .03). Homeless patients with advanced NSCLC had a median survival of 0.58 years, versus 1.30 years in housed patients ( P = .48)., Conclusion: To our knowledge, this is the first US study comparing outcomes among homeless and housed patients with NSCLC within the same institution; we found homeless patients had longer delays to biopsy, increased rates of missed appointments, and a trend toward decreased survival. This study shows potential areas where interventions could be implemented to improve lung cancer outcomes in this patient population.
- Published
- 2020
- Full Text
- View/download PDF
17. A Phase II Trial of Pembrolizumab and Vorinostat in Recurrent Metastatic Head and Neck Squamous Cell Carcinomas and Salivary Gland Cancer.
- Author
-
Rodriguez CP, Wu QV, Voutsinas J, Fromm JR, Jiang X, Pillarisetty VG, Lee SM, Santana-Davila R, Goulart B, Baik CS, Chow LQM, Eaton K, and Martins R
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Drug Resistance, Neoplasm, Female, Head and Neck Neoplasms pathology, Humans, Male, Middle Aged, Neoplasm Metastasis, Neoplasm Recurrence, Local pathology, Salivary Gland Neoplasms pathology, Squamous Cell Carcinoma of Head and Neck pathology, Survival Rate, Vorinostat administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Head and Neck Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Salivary Gland Neoplasms drug therapy, Squamous Cell Carcinoma of Head and Neck drug therapy
- Abstract
Purpose: This clinical trial combined pembrolizumab and vorinostat in recurrent/metastatic squamous cell carcinomas of the head and neck (HN), and salivary gland cancer (SGC)., Patients and Methods: Patients with progressing incurable HN and SGC, Eastern Cooperative Oncology Group (ECOG) ≤1, no prior immunotherapy, RECIST1.1 measurable disease, and normal organ function were eligible. Pembrolizumab 200 mg was given intravenous every 21 days, and vorinostat 400 mg given orally 5 days on and 2 days off during each 21-day cycle. Primary endpoints were safety and objective response rates., Results: From November 2015 to August 2017, 25 patients with HN and 25 SGC were enrolled. Median age was 61 (range, 33-86) years, 39 (78%) were male, 21 (62%) were never smokers, and 27 (54%) had ECOG 0. In HN, 13 (52%) were p16+ oropharynx. Most common SGC histologies were adenoid cystic 12 (48%), acinic cell 3 (12%), and mucoepidermoid 3 (12%). Adverse events (AEs) in all patients were: 27 (54%) with grade ≥ 1 and 18 (36%) with grade ≥ 3. The most common AEs in all patients were renal insufficiency in seven, (14%), fatigue in six, (12%), and nausea in three (6%). There were three (12%) deaths on study. Responses in HN were complete response (CR) 0, partial response (PR) eight (32%), and stable disease (SD) five (20%). Efficacy in SGCs was CR 0, PR four (16%) in one lymphoepithelioma-like carcinoma, two acinic cell, one adenoid cystic, and SD 14 (56%). In the HN group, median follow-up (mFUP) was 12.6 months, median overall survival (mOS) was 12.6 months, and median progression-free survival (mPFS) was 4.5 months. In SGC, mFUP was 13.1 months, mOS was 14.0 months, and mPFS was 6.9 months., Conclusions: This combination demonstrated activity in HN, with fewer responses in SGC. Toxicities were higher than reported with pembrolizumab alone., (©2019 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
18. Validity of Natural Language Processing for Ascertainment of EGFR and ALK Test Results in SEER Cases of Stage IV Non-Small-Cell Lung Cancer.
- Author
-
Goulart BHL, Silgard ET, Baik CS, Bansal A, Sun Q, Durbin EB, Hands I, Shah D, Arnold SM, Ramsey SD, Kavuluru R, and Schwartz SM
- Subjects
- Adult, Aged, Algorithms, Carcinoma, Non-Small-Cell Lung epidemiology, DNA Mutational Analysis, ErbB Receptors genetics, Female, Genetic Testing, Humans, Kentucky epidemiology, Lung Neoplasms epidemiology, Machine Learning, Male, Middle Aged, Population Surveillance, Registries, Reproducibility of Results, SEER Program, Anaplastic Lymphoma Kinase genetics, Carcinoma, Non-Small-Cell Lung diagnosis, Carcinoma, Non-Small-Cell Lung etiology, Lung Neoplasms diagnosis, Lung Neoplasms etiology, Mutation, Natural Language Processing
- Abstract
Purpose: SEER registries do not report results of epidermal growth factor receptor ( EGFR ) and anaplastic lymphoma kinase ( ALK ) mutation tests. To facilitate population-based research in molecularly defined subgroups of non-small-cell lung cancer (NSCLC), we assessed the validity of natural language processing (NLP) for the ascertainment of EGFR and ALK testing from electronic pathology (e-path) reports of NSCLC cases included in two SEER registries: the Cancer Surveillance System (CSS) and the Kentucky Cancer Registry (KCR)., Methods: We obtained 4,278 e-path reports from 1,634 patients who were diagnosed with stage IV nonsquamous NSCLC from September 1, 2011, to December 31, 2013, included in CSS. We used 855 CSS reports to train NLP systems for the ascertainment of EGFR and ALK test status (reported v not reported) and test results (positive v negative). We assessed sensitivity, specificity, and positive and negative predictive values in an internal validation sample of 3,423 CSS e-path reports and repeated the analysis in an external sample of 1,041 e-path reports from 565 KCR patients. Two oncologists manually reviewed all e-path reports to generate gold-standard data sets., Results: NLP systems yielded internal validity metrics that ranged from 0.95 to 1.00 for EGFR and ALK test status and results in CSS e-path reports. NLP showed high internal accuracy for the ascertainment of EGFR and ALK in CSS patients-F scores of 0.95 and 0.96, respectively. In the external validation analysis, NLP yielded metrics that ranged from 0.02 to 0.96 in KCR reports and F scores of 0.70 and 0.72, respectively, in KCR patients., Conclusion: NLP is an internally valid method for the ascertainment of EGFR and ALK test information from e-path reports available in SEER registries, but future work is necessary to increase NLP external validity.
- Published
- 2019
- Full Text
- View/download PDF
19. Real-world treatment patterns and survival of patients with BRAF V600-mutated metastatic non-small cell lung cancer.
- Author
-
Horn L, Bauml J, Forde PM, Davis KL, Myall NJ, Sasane M, Dalal A, Culver K, Wozniak AJ, Baik CS, Mutebi A, Zhang P, Wakelee HA, and Johnson BE
- Subjects
- Adult, Aged, Aged, 80 and over, Biomarkers, Tumor, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung therapy, Clinical Decision-Making, Combined Modality Therapy, Cross-Sectional Studies, Disease Management, Female, Humans, Lung Neoplasms therapy, Male, Middle Aged, Neoplasm Metastasis, Neoplasm Staging, Prognosis, Retrospective Studies, Survival Analysis, Treatment Outcome, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung mortality, Lung Neoplasms genetics, Lung Neoplasms mortality, Mutation, Practice Patterns, Physicians', Proto-Oncogene Proteins B-raf genetics
- Abstract
Introduction: Clinical outcomes data on BRAF-mutated non-small cell lung cancer (NSCLC) patients treated in routine practice is limited. To address this gap, we described treatment patterns and survival in a cohort of these patients evaluated/treated at 7 US academic cancer centers during 2009-2016., Methods: This was a retrospective chart review. Patients with BRAF V600-mutated metastatic NSCLC were selected. Current/previous participants in BRAF-related trials were excluded. Onset of metastatic NSCLC defined a patient's index date, which had to occur ≥6 months before the chart review date. Analyses were descriptive, including Kaplan-Meier analyses for overall survival (OS)., Results: The study included 72 patients. At index, median age (range) was 65 (44-90) years; 61.1% were female. Fifty-two patients received ≥1 line of systemic therapy for metastatic disease. Platinum-based doublet chemotherapy was the most common first-line (1 L) regimen (76.9% of 1 l recipients); no patient received 1 l targeted therapy (TT) with a BRAF/MEK inhibitor. In total, 20 patients received TT in any treatment line (2 l or later). At time of review, 38 patients were deceased. Median (95%CI) OS from index for all patients was 31.0 (14.5, 63.8) months. Median (95%CI) OS was 56.5 (13.4, 89.1) months from index for TT recipients and 27.2 (10.6, 64.6) months in patients not treated with TT., Conclusion: Survival time in BRAF V600-mutated metastatic NSCLC patients studied here was higher than expected based on indirect comparisons with historical NSCLC cohorts for whom no oncogenic driver (BRAF or otherwise) was present. TT recipients had a numerically longer OS from metastatic onset than patients receiving usual care, further highlighting the importance of TT in BRAF V600-mutant NSCLC., (Copyright © 2018 Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
20. Trastuzumab Emtansine (T-DM1) in Patients with Previously Treated HER2-Overexpressing Metastatic Non-Small Cell Lung Cancer: Efficacy, Safety, and Biomarkers.
- Author
-
Peters S, Stahel R, Bubendorf L, Bonomi P, Villegas A, Kowalski DM, Baik CS, Isla D, Carpeno JC, Garrido P, Rittmeyer A, Tiseo M, Meyenberg C, de Haas S, Lam LH, Lu MW, and Stinchcombe TE
- Subjects
- Ado-Trastuzumab Emtansine adverse effects, Adult, Aged, Aged, 80 and over, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Disease-Free Survival, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Neoplasm Staging, Ado-Trastuzumab Emtansine administration & dosage, Biomarkers, Tumor genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Receptor, ErbB-2 genetics
- Abstract
Purpose: HER2-targeted therapy is not standard of care for HER2-positive non-small cell lung cancer (NSCLC). This phase II study investigated efficacy and safety of the HER2-targeted antibody-drug conjugate trastuzumab emtansine (T-DM1) in patients with previously treated advanced HER2-overexpressing NSCLC., Patients and Methods: Eligible patients had HER2-overexpressing NSCLC (centrally tested IHC) and received previous platinum-based chemotherapy and targeted therapy in the case of EGFR mutation or ALK gene rearrangement. Patients were divided into cohorts based on HER2 IHC (2+, 3+). All patients received T-DM1 3.6 mg/kg intravenously every 3 weeks until disease progression or unacceptable toxicity. The primary endpoint was investigator-determined overall response rate (ORR) using RECIST v1.1., Results: Forty-nine patients received T-DM1 (29 IHC 2+, 20 IHC 3+). No treatment responses were observed in the IHC 2+ cohort. Four partial responses were observed in the IHC 3+ cohort (ORR, 20%; 95% confidence interval, 5.7%-43.7%). Clinical benefit rates were 7% and 30% in the IHC 2+ and 3+ cohorts, respectively. Response duration for the responders was 2.9, 7.3, 8.3, and 10.8 months. Median progression-free survival and overall survival were similar between cohorts. Three of 4 responders had HER2 gene amplification. No new safety signals were observed., Conclusions: T-DM1 showed a signal of activity in patients with HER2-overexpressing (IHC 3+) advanced NSCLC. Additional investigation into HER2 pathway alterations is needed to refine the target population for T-DM1 in NSCLC; however, HER2 IHC as a single parameter was an insufficient predictive biomarker., (©2018 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
21. Immuno-oncology Clinical Trial Design: Limitations, Challenges, and Opportunities.
- Author
-
Baik CS, Rubin EH, Forde PM, Mehnert JM, Collyar D, Butler MO, Dixon EL, and Chow LQM
- Subjects
- Humans, Neoplasms immunology, Research Design, Clinical Trials as Topic, Medical Oncology trends, Neoplasms drug therapy
- Abstract
Recent advances in immuno-oncology and regulatory approvals have been rapid and paradigm shifting in many difficult-to-treat malignancies. Despite immune checkpoint inhibitor therapy becoming the standard of care across multiple tumor types, there are many unanswered questions that need to be addressed before this therapeutic modality can be fully harnessed. Areas of limitations include treatment of patients not sufficiently represented in clinical trials, uncertainty of the optimal treatment dosing and duration, and lack of understanding regarding long-term immune related toxicities and atypical tumor responses. Patients such as those with autoimmune disease, chronic viral infections, limited performance status, and brain metastases were often excluded from initial trials due to concerns of safety. However, limited data suggest that some of these patients can benefit from therapy with manageable toxicities; thus, future studies should incorporate these patients to clearly define safety and efficacy. There are still controversies regarding the optimal dosing strategy that can vary from weight-based to flat dosing, with undefined treatment duration. Further elucidation of the optimal dosing approach and evaluation of predictive biomarkers should be incorporated in the design of future trials. Finally, there are long-term immune-mediated toxicities, atypical tumor responses such as pseudoprogression and endpoints unique to immuno-oncology that are not adequately captured by traditional trial designs; thus, novel study designs are needed. In this article, we discuss in detail the above challenges and propose needed areas of research for exploration and incorporation in the next generation of immuno-oncology clinical trials., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
22. Predictors of outcome with cetuximab and paclitaxel for head and neck squamous cell carcinoma.
- Author
-
Pellini Ferreira B, Redman M, Baker KK, Martins R, Eaton KD, Chow LQM, Baik CS, Goulart B, Lee SM, Santana-Davila R, and Rodriguez CP
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Squamous Cell mortality, Carcinoma, Squamous Cell pathology, Cetuximab adverse effects, Disease-Free Survival, Drug Administration Schedule, Female, Follow-Up Studies, Humans, Male, Middle Aged, Neoplasm Recurrence, Local drug therapy, Neoplasm Recurrence, Local pathology, Otorhinolaryngologic Neoplasms mortality, Paclitaxel adverse effects, Prognosis, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Squamous Cell drug therapy, Cetuximab administration & dosage, Otorhinolaryngologic Neoplasms drug therapy, Paclitaxel administration & dosage
- Abstract
Objectives: Identify predictors of outcome in patients with recurrent/metastatic head and neck squamous cell carcinoma (RMHNSCC) treated with weekly cetuximab and paclitaxel (CP)., Study Design: Retrospective analysis., Methods: Patients with RMHNSCC treated with CP were identified and patient data was recorded. The Kaplan-Meier method was used to estimate outcomes, and Cox regression analysis was used to examine outcome predictors., Results: Fifty-nine patients initiated CP between January 2007 and June 2014. Median age was 56 (range: 39-80) years. The most common primary sites were the oropharynx in 22 (37%) patients, oral cavity in 19 (32%), and larynx in 11 (19%). Eastern Cooperative Oncology Group performance status (ECOG PS) was 0 in seven (12%), 1 in 32 (54%), and 2 in 16 (28%) patients. In 44 (75%) patients, CP was used as a first-line R/M regimen. Median number of cycles was five (range: 1-29). Dose modifications were necessary in 27 (46%) patients. The objective response rate was 47.5%, with 27 (45.8%) partial responses and one (2%) complete response. With a median follow-up of 13.4 months, median progression-free (PFS) and overall survival (OS) were 7.7 and 13.2 months, respectively. On multivariable analysis, an ECOG of 2 of 3 was associated with inferior OS (hazard ratio [HR]: 3.94; P = 0.01; 95% confidence interval [CI]: 1.1-14.04) and PFS (HR: 7.29; P < 0.01; 95% CI: 2.1-26.0) compared to an ECOG 0 of 1. First-line CP administration was associated with superior PFS compared to second line (HR: 2.6; P = 0.02; 95% CI:1.2-5.5)., Conclusions: CP is well tolerated in this population of patients, with favorable tumor efficacy. First-line use and an ECOG 0 of 1 points appears to confer superior outcomes., Level of Evidence: 4. Laryngoscope, 127:1583-1588, 2017., (© 2016 The American Laryngological, Rhinological and Otological Society, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
23. Targeting BRAF -Mutant Non-Small Cell Lung Cancer: From Molecular Profiling to Rationally Designed Therapy.
- Author
-
Baik CS, Myall NJ, and Wakelee HA
- Subjects
- Antineoplastic Agents pharmacology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Drug Resistance, Neoplasm drug effects, Drug Resistance, Neoplasm genetics, Humans, Lung Neoplasms genetics, Lung Neoplasms pathology, Proto-Oncogene Mas, Proto-Oncogene Proteins B-raf antagonists & inhibitors, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Molecular Targeted Therapy methods, Mutation, Proto-Oncogene Proteins B-raf genetics
- Abstract
Non-small cell lung cancer (NSCLC) remains the leading cause of cancer-related deaths globally. However, the identification of oncogenic driver alterations involved in the initiation and maintenance of NSCLC, such as epidermal growth factor receptor mutations and anaplastic lymphoma kinase translocation, has led to the development of novel therapies that directly target mutant proteins and associated signaling pathways, resulting in improved clinical outcomes. As sequencing techniques have improved, the molecular heterogeneity of NSCLC has become apparent, leading to the identification of a number of potentially actionable oncogenic driver mutations. Of these, one of the most promising therapeutic targets is B-Raf proto-oncogene, serine/threonine kinase (BRAF). Mutations in BRAF, observed in 2%-4% of NSCLCs, typically lead to constitutive activation of the protein and, as a consequence, lead to activation of the mitogen-activated protein kinase signaling pathway. Direct inhibition of mutant BRAF and/or the downstream mitogen-activated protein kinase kinase (MEK) has led to prolonged survival in patients with BRAF -mutant metastatic melanoma. This comprehensive review will discuss the clinical characteristics and prognostic implications of BRAF -mutant NSCLC, the clinical development of BRAF and MEK inhibitors from melanoma to NSCLC, and practical considerations for clinicians involving BRAF mutation screening and the choice of targeted therapy., Implications for Practice: Personalized medicine has begun to provide substantial benefit to patients with oncogene-driven non-small cell lung cancer (NSCLC). However, treatment options for patients with oncogenic driver mutations lacking targeted treatment strategies remain limited. Direct inhibition of mutant B-Raf proto-oncogene, serine/threonine kinase (BRAF) and/or downstream mitogen-activated protein kinase kinase (MEK) has the potential to change the course of the disease for patients with BRAF -mutant NSCLC, as it has in BRAF -mutant melanoma. Optimization of screening strategies for rare mutations and the choice of appropriate agents on an individual basis will be key to providing timely and successful intervention., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© 2017 The Authors. The Oncologist published by Wiley Periodicals, Inc. on behalf of AlphaMed Press.)
- Published
- 2017
- Full Text
- View/download PDF
24. Phase Ib Trial of the Toll-like Receptor 8 Agonist, Motolimod (VTX-2337), Combined with Cetuximab in Patients with Recurrent or Metastatic SCCHN.
- Author
-
Chow LQM, Morishima C, Eaton KD, Baik CS, Goulart BH, Anderson LN, Manjarrez KL, Dietsch GN, Bryan JK, Hershberg RM, Disis ML, and Martins RG
- Subjects
- Adult, Aged, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Benzazepines adverse effects, Carcinoma, Squamous Cell blood, Carcinoma, Squamous Cell genetics, Cetuximab administration & dosage, Disease-Free Survival, Drug-Related Side Effects and Adverse Reactions classification, Drug-Related Side Effects and Adverse Reactions pathology, Female, Head and Neck Neoplasms blood, Head and Neck Neoplasms genetics, Humans, Killer Cells, Natural metabolism, Male, Middle Aged, Neoplasm Metastasis, Neoplasm Recurrence, Local pathology, Squamous Cell Carcinoma of Head and Neck, Benzazepines administration & dosage, Carcinoma, Squamous Cell drug therapy, Head and Neck Neoplasms drug therapy, Neoplasm Recurrence, Local drug therapy, Toll-Like Receptor 8 genetics
- Abstract
Purpose: As Toll-like receptors (TLR) are key mediators of immune responses, TLR agonists may be important for augmenting the efficacy of therapies for squamous cell carcinoma of the head and neck (SCCHN). Motolimod (VTX-2337), a selective small-molecule agonist of TLR8, stimulates natural killer (NK) cells, dendritic cells, and monocytes. A phase Ib clinical trial assessed the safety and antitumor activity of motolimod in combination with cetuximab in patients with SCCHN. Correlative biomarkers of immune activity were explored. Experimental Design: Thirteen patients with recurrent or metastatic SCCHN were enrolled in this open-label, dose-escalation study using a standard 3 + 3 design. Doses of motolimod (2.5, 3.0, or 3.5 mg/m
2 ) were given on days 1, 8, and 15, in combination with fixed weekly doses of cetuximab in 28-day cycles. Results: There were no protocol-defined dose-limiting toxicities, drug-related deaths, or evidence of synergistic toxicities between motolimod and cetuximab. Clinical tolerability at the 3.5 mg/m2 dose level was not optimal for repeated dosing and 3.0 mg/m2 was identified as the MTD. Two patients achieved partial responses for an overall response rate of 15%. Five patients had disease stabilization equating to a disease control rate of 54%. Statistically significant increases in plasma cytokines and in the frequency and activation of circulating NK cells were observed. Conclusions: Motolimod can be safely administered in combination with cetuximab with an acceptable toxicity profile. Encouraging antitumor activity and robust pharmacodynamic responses were observed. Motolimod is being further investigated in a phase II trial in patients with SCCHN (ClinicalTrials.gov ID: NCT01836029). Clin Cancer Res; 23(10); 2442-50. ©2016 AACR ., (©2016 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
25. Systemic therapy of brain metastases: non-small cell lung cancer, breast cancer, and melanoma.
- Author
-
Chamberlain MC, Baik CS, Gadi VK, Bhatia S, and Chow LQ
- Subjects
- Brain Neoplasms secondary, Breast Neoplasms pathology, Carcinoma, Non-Small-Cell Lung pathology, Female, Humans, Lung Neoplasms pathology, Melanoma pathology, Molecular Targeted Therapy, Brain Neoplasms therapy, Breast Neoplasms therapy, Carcinoma, Non-Small-Cell Lung therapy, Lung Neoplasms therapy, Melanoma therapy
- Abstract
Brain metastases (BM) occur frequently in many cancers, particularly non-small cell lung cancer (NSCLC), breast cancer, and melanoma. The development of BM is associated with poor prognosis and has an adverse impact on survival and quality of life. Commonly used therapies for BM such as surgery or radiotherapy are associated with only modest benefits. However, recent advances in systemic therapy of many cancers have generated considerable interest in exploration of those therapies for treatment of intracranial metastases.This review discusses the epidemiology of BM from the aforementioned primary tumors and the challenges of using systemic therapies for metastatic disease located within the central nervous system. Cumulative data from several retrospective and small prospective studies suggest that molecularly targeted systemic therapies may be an effective option for the treatment of BM from NSCLC, breast cancer, and melanoma, either as monotherapy or in conjunction with other therapies. Larger prospective studies are warranted to further characterize the efficacy and safety profiles of these targeted agents for the treatment of BM., (© The Author(s) 2016. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
26. Dabrafenib plus trametinib in patients with previously treated BRAF(V600E)-mutant metastatic non-small cell lung cancer: an open-label, multicentre phase 2 trial.
- Author
-
Planchard D, Besse B, Groen HJM, Souquet PJ, Quoix E, Baik CS, Barlesi F, Kim TM, Mazieres J, Novello S, Rigas JR, Upalawanna A, D'Amelio AM Jr, Zhang P, Mookerjee B, and Johnson BE
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Adenocarcinoma secondary, Adult, Aged, Biomarkers, Tumor genetics, Carcinoma, Large Cell drug therapy, Carcinoma, Large Cell genetics, Carcinoma, Large Cell secondary, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung secondary, Female, Follow-Up Studies, Humans, Imidazoles administration & dosage, Lung Neoplasms genetics, Lung Neoplasms pathology, Lymphatic Metastasis, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Neoplasm Staging, Oximes administration & dosage, Prognosis, Pyridones administration & dosage, Pyrimidinones administration & dosage, Survival Rate, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Mutation genetics, Neoplasm Recurrence, Local drug therapy, Proto-Oncogene Proteins B-raf genetics
- Abstract
Background: BRAF mutations act as an oncogenic driver via the mitogen-activated protein kinase (MAPK) pathway in non-small cell lung cancer (NSCLC). BRAF inhibition has shown antitumour activity in patients with BRAF(V600E)-mutant NSCLC. Dual MAPK pathway inhibition with BRAF and MEK inhibitors in BRAF(V600E)-mutant NSCLC might improve efficacy over BRAF inhibitor monotherapy based on observations in BRAF(V600)-mutant melanoma. We aimed to assess the antitumour activity and safety of dabrafenib plus trametinib in patients with BRAF(V600E)-mutant NSCLC., Methods: In this phase 2, multicentre, non-randomised, open-label study, we enrolled adult patients (aged ≥18 years) with pretreated metastatic stage IV BRAF(V600E)-mutant NSCLC who had documented tumour progression after at least one previous platinum-based chemotherapy and had had no more than three previous systemic anticancer therapies. Patients with previous BRAF or MEK inhibitor treatment were ineligible. Patients with brain metastases were allowed to enrol only if the lesions were asymptomatic, untreated (or stable more than 3 weeks after local therapy if treated), and measured less than 1 cm. Enrolled patients received oral dabrafenib (150 mg twice daily) plus oral trametinib (2 mg once daily) in continuous 21-day cycles until disease progression, unacceptable adverse events, withdrawal of consent, or death. The primary endpoint was investigator-assessed overall response, which was assessed by intention to treat in the protocol-defined population (patients who received second-line or later treatment); safety was also assessed in this population and was assessed at least once every 3 weeks, with adverse events, laboratory values, and vital signs graded according to the Common Terminology Criteria for Adverse Events version 4.0. The study is ongoing but no longer recruiting patients. This trial is registered with ClinicalTrials.gov, number NCT01336634., Findings: Between Dec 20, 2013, and Jan 14, 2015, 59 patients from 30 centres in nine countries across North America, Europe, and Asia met eligibility criteria. Two patients who had previously been untreated due to protocol deviation were excluded; thus, 57 eligible patients were enrolled. 36 patients (63·2% [95% CI 49·3-75·6]) achieved an investigator-assessed overall response. Serious adverse events were reported in 32 (56%) of 57 patients and included pyrexia in nine (16%), anaemia in three (5%), confusional state in two (4%), decreased appetite in two (4%), haemoptysis in two (4%), hypercalcaemia in two (4%), nausea in two (4%), and cutaneous squamous cell carcinoma in two (4%). The most common grade 3-4 adverse events were neutropenia in five patients (9%), hyponatraemia in four (7%), and anaemia in three (5%). Four patients died during the study from fatal adverse events judged to be unrelated to treatment (one retroperitoneal haemorrhage, one subarachnoid haemorrhage, one respiratory distress, and one from disease progression that was more severe than typical progression, as assessed by the investigator)., Interpretation: Dabrafenib plus trametinib could represent a new targeted therapy with robust antitumour activity and a manageable safety profile in patients with BRAF(V600E)-mutant NSCLC., Funding: GlaxoSmithKline., Competing Interests: Declaration of interests DP acts as an advisor for AstraZeneca, Boehringer, Bristol-Myers Squibb, Clovis, Lilly, Merck Sharp & Dohme, Novartis, Pfizer, Roche, and Sanofi and has received research funding from Novartis unrelated to the current trial. BB’s institution received a grant from Novartis for this study. HJMG’s institution has received payments from Eli Lilly, Merck Sharp & Dohme, and Roche. P-JS has been involved in clinical trials for GlaxoSmithKline and Novartis. EQ has received personal fees from AbbVie, Bristol-Myers Squibb, Clovis, Lilly, Pfizer, and Roche, has received grants from Amgen, Boehringer, Bristol-Myers Squibb, Lilly, Merck Sharp & Dohme, Mundipharma, and Roche, and has received non-financial support from Boehringer. CSB’s institution received a grant for this study from Novartis and CSB has received personal fees from Novartis. FB has received personal fees from Novartis. SN has received personal fees from Boehringer, Bristol-Myers Squibb, Eli Lilly, Merck Sharp & Dohme, and Roche. AU, PZ, AD’A, and BM are currently employees of Novartis. AD’A and BM were employees of GlaxoSmithKline during a portion of the study. AD’A and BM own stock in GlaxoSmithKline and Novartis and BM owns stock in Incyte and AstraZeneca. BEJ has received personal fees from Amgen, AstraZeneca, Boehringer, Chugai Pharmaceuticals, Clovis, Genentech, KEW Group, Merck, and Novartis, and shares of post-market revenue for an EGFR genotyping patent. The other authors declare no competing interests., (Copyright © 2016 Elsevier Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
27. Adjuvant Radiotherapy for Stages II and III Resected Thymoma: A Single-institutional Experience.
- Author
-
Yan J, Liu Q, Moseley JN, Baik CS, Chow LQ, Goulart BH, Zlotnick D, Papanicolau-Sengos A, Gallaher I, Knopp JM, Zeng J, and Patel S
- Subjects
- Disease-Free Survival, Female, Humans, Male, Margins of Excision, Middle Aged, Neoplasm Invasiveness, Neoplasm Staging, Neoplasm, Residual, Radiotherapy, Adjuvant, Retrospective Studies, Survival Rate, Thymoma surgery, Thymus Neoplasms surgery, Thymoma pathology, Thymoma radiotherapy, Thymus Neoplasms pathology, Thymus Neoplasms radiotherapy
- Abstract
Introduction: The role of adjuvant radiation for Masaoka stages II and III thymoma remains controversial. The aim of this study was to evaluate the clinical benefit of radiation therapy for resected stages II and III thymoma patients., Methods: We retrospectively reviewed the medical records of 175 thymoma patients treated from July 1996 to January 2013 at University of Washington Medical Center; 88 patients with adequate follow-up and who met histologic criteria were included. We evaluated progression-free survival (PFS) and overall survival (OS), and compared these outcomes in patients treated by surgery (S) alone versus surgery plus radiotherapy (S+RT). Cox regression models and log-rank tests were used to compare PFS and OS for S versus S+RT, and they were further assessed by margin-positive versus margin-negative subgroups using Kaplan-Meier curves., Results: Among the 88 thymoma patients, 22 were stage II and 18 were stage III. For all stages II and III patients, adjuvant radiation was not identified as a significant predictor for PFS (P=0.95) or OS (P=0.63). A positive surgical margin predicted for a worse OS (hazard ratio=7.1; P=0.004). Further investigation revealed for resection margin-positive patients; S+RT had higher OS than S alone (P=0.006)., Conclusions: For stages II and III thymoma, postoperative adjuvant radiation was not associated with statistically significant differences in PFS or OS in this study. Our results indicated a potential OS benefit of adjuvant RT in patients with positive resection margins, and therefore may be considered in this patient population.
- Published
- 2016
- Full Text
- View/download PDF
28. Durable Response to Tyrosine Kinase Inhibitor Therapy in a Lung Cancer Patient Harboring Epidermal Growth Factor Receptor Tandem Kinase Domain Duplication.
- Author
-
Baik CS, Wu D, Smith C, Martins RG, and Pritchard CC
- Subjects
- ErbB Receptors metabolism, Female, Humans, Lung Neoplasms enzymology, Middle Aged, Protein Structure, Tertiary, Carcinoma, Non-Small-Cell Lung drug therapy, ErbB Receptors antagonists & inhibitors, Lung Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use
- Published
- 2015
- Full Text
- View/download PDF
29. Targeted Therapy for Brain Metastases in EGFR-Mutated and ALK-Rearranged Non-Small-Cell Lung Cancer.
- Author
-
Baik CS, Chamberlain MC, and Chow LQ
- Subjects
- Anaplastic Lymphoma Kinase, Brain Neoplasms genetics, Carcinoma, Non-Small-Cell Lung pathology, ErbB Receptors metabolism, Humans, Lung Neoplasms pathology, Brain Neoplasms secondary, Carcinoma, Non-Small-Cell Lung genetics, ErbB Receptors genetics, Lung Neoplasms genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
Approximately half of all patients with non-small-cell lung cancer (NSCLC) develop brain metastases (BM) during the course of their disease, leading to significant challenges in treatment. Molecular targeted tyrosine kinase inhibitors have proven effective for patients with activating mutations in the epidermal growth factor receptor gene and chromosomal rearrangements involving the anaplastic lymphoma kinase gene. Despite their efficacy in systemic disease control, their effectiveness in patients with BM is not well established. In this article, we review recent data on the use of epidermal growth factor receptor and anaplastic lymphoma kinase tyrosine kinase inhibitors for treatment of patients with NSCLC and BM. These data highlight the potential for meaningful disease control within the central nervous system and the inherent challenges in treating patients with NSCLC and BM.
- Published
- 2015
- Full Text
- View/download PDF
30. Nonsteroidal Anti-Inflammatory Drug and Aspirin Use in Relation to Lung Cancer Risk among Postmenopausal Women.
- Author
-
Baik CS, Brasky TM, Pettinger M, Luo J, Gong Z, Wactawski-Wende J, and Prentice RL
- Subjects
- Aged, Female, Humans, Incidence, Middle Aged, Proportional Hazards Models, Risk Factors, Anti-Inflammatory Agents, Non-Steroidal therapeutic use, Aspirin therapeutic use, Lung Neoplasms epidemiology, Postmenopause
- Abstract
Background: Results from prospective studies suggest that nonsteroidal anti-inflammatory drugs (NSAID) may decrease lung cancer risk; however, any protective effect appears to be most evident in men., Methods: We evaluated the associations between NSAID use and lung cancer incidence in postmenopausal women in the Women's Health Initiative (WHI) adjusting for female-specific potential confounders such as hormone therapy in addition to smoking histories and other potential confounders. We identified 143,841 women from ages 50 to 79 and 1,902 centrally confirmed lung cancer cases were included in the analysis. We used Cox regression models to estimate HRs and their 95% confidence intervals (CI)., Results: Compared with nonuse, regular NSAID use was not associated with overall lung cancer incidence (NSAID use >10 years HR 0.87; 95% CI, 0.71-1.08, P(trend) = 0.13). No statistically significant associations were found when examined by histologic subtypes and although there was a trend of decreased risk with longer duration of NSAID use in the adenocarcinoma subtype, this was not statistically significant (NSAID use >10 years HR 0.80; 95% CI, 0.58-1.10; P(trend) = 0.07)., Conclusion: Our study did not show that NSAID use is associated with lung cancer risk in women even after adjusting for female-specific confounders. There was a trend of decreased risk in the adenocarcinoma subtype; however, this was not statistically significant., Impact: Future studies will need to take in account the various molecular subtypes of non-small cell lung cancer to further elucidate the role of NSAIDs in lung cancer, especially for the adenocarcinoma subtype., (©2015 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
31. Non-steroidal anti-inflammatory drugs and cancer risk in women: results from the Women's Health Initiative.
- Author
-
Brasky TM, Liu J, White E, Peters U, Potter JD, Walter RB, Baik CS, Lane DS, Manson JE, Vitolins MZ, Allison MA, Tang JY, and Wactawski-Wende J
- Subjects
- Aged, Female, Humans, Incidence, Middle Aged, Neoplasms epidemiology, Postmenopause, Prospective Studies, Risk, Anti-Inflammatory Agents, Non-Steroidal administration & dosage, Anticarcinogenic Agents administration & dosage, Aspirin administration & dosage, Ibuprofen administration & dosage, Naproxen administration & dosage, Neoplasms prevention & control
- Abstract
The use of non-steroidal anti-inflammatory drugs (NSAIDs) has been associated with reduced risks of cancers at several sites in some studies; however, we recently reported no association between their use and total cancer risk in women in a prospective study. Here we examine the association between NSAIDs and total and site-specific cancer incidence in the large, prospective Women's Health Initiative (WHI). Women (129,013) were recruited to participate in the WHI at 40 US clinical centers from 1993 to 1998 and followed prospectively. After 9.7 years of follow-up, 12,998 incident, first primary, invasive cancers were diagnosed. NSAID use was systematically collected at study visits. We used Cox proportional hazards regression models to estimate multivariable-adjusted hazard ratios (HR) and 95% confidence intervals (CI) for associations between NSAIDs use and total and site-specific cancer risk. Relative to non-use, consistent use (i.e., use at baseline and year 3 of follow-up) of any NSAID was not associated with total cancer risk (HR 1.00, 95% CI: 0.94-1.06). Results for individual NSAIDs were similar to the aggregate measure. In site-specific analyses, NSAIDs were associated with reduced risks of colorectal cancer, ovarian cancer, and melanoma. Our study confirms a chemopreventive benefit for colorectal cancer in women and gives preliminary evidence for a reduction of the risk of some rarer cancers. NSAIDs' benefit on cancer risk was therefore limited to specific sites and not evident when total cancer risk was examined. This information may be of importance when NSAIDs are considered as chemopreventive agents., (© 2014 UICC.)
- Published
- 2014
- Full Text
- View/download PDF
32. The role of chemotherapy in the management of stage IIIA non-small cell lung cancer.
- Author
-
Baik CS, Vallières E, and Martins RG
- Subjects
- Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung therapy, Chemoradiotherapy, Humans, Induction Chemotherapy, Lung Neoplasms pathology, Lung Neoplasms therapy, Lymph Nodes pathology, Neoadjuvant Therapy, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy
- Abstract
Patients with confirmed stage IIIA non-small cell lung cancer (NSCLC) represent a very heterogeneous group which includes those with limited microscopic ipsilateral mediastinal lymph node involvement discovered after a surgical resection, as well as those who have radiologically evident bulky subcarinal lymph node involvement at presentation. Different therapeutic options in stage IIIA disease include neoadjuvant chemo- or chemoradiotherapy followed by surgery, primary surgery followed by adjuvant chemotherapy with or without sequential adjuvant radiation therapy or definitive chemoradiation without surgery. The roles of surgery and radiation in stage IIIA disease are controversial, and there is inadequate data from randomized trials to inform the optimal therapeutic strategy. In contrast, chemotherapy has a clear indication in the curative setting. Data from randomized trials indicates that cisplatin-based chemotherapy should be given in either adjuvant or neoadjuvant settings to patients who are undergoing curative surgical resection and who are candidates for cisplatin therapy. In definitive chemoradiotherapy, cisplatin-based therapy is recommended although a carboplatin-based regimen may be given if patients cannot receive cisplatin. Finally, all patients with stage IIIA NSCLC should be evaluated early in a multidisciplinary setting that includes medical and radiation oncologists and thoracic surgeons with experience in lung cancer therapy.
- Published
- 2013
- Full Text
- View/download PDF
33. Estrogen signaling in lung cancer: an opportunity for novel therapy.
- Author
-
Baik CS and Eaton KD
- Abstract
Lung cancer is the leading cause of cancer death in U.S. and represents a major public health burden. Epidemiologic data have suggested that lung cancer in women may possess different biological characteristics compared to men, as evidenced by a higher proportion of never-smokers among women with lung cancer. Emerging data indicate that female hormones such as estrogen and progesterone play a significant role in lung carcinogenesis. It has been reported that estrogen and progesterone receptors are expressed in lung cancer cell lines as well as in patient-derived tumors. Hormone related risk factors such as hormone replacement therapy have been implicated in lung carcinogenesis and several preclinical studies show activity of anti-estrogen therapy in lung cancer. In this review, we summarize the emerging evidence for the role of reproductive hormones in lung cancer and implications for lung cancer therapy.
- Published
- 2012
- Full Text
- View/download PDF
34. Non-steroidal anti-inflammatory drugs and small cell lung cancer risk in the VITAL study.
- Author
-
Brasky TM, Baik CS, Slatore CG, Potter JD, and White E
- Subjects
- Aged, Female, Humans, Lung Neoplasms epidemiology, Male, Middle Aged, Risk Assessment, Risk Factors, Small Cell Lung Carcinoma epidemiology, Surveys and Questionnaires, Washington epidemiology, Anti-Inflammatory Agents, Non-Steroidal adverse effects, Lung Neoplasms etiology, Small Cell Lung Carcinoma etiology
- Abstract
Few studies have examined the association between non-steroidal anti-inflammatory drug (NSAID) use and risk of small cell lung cancer (SCLC); among them, findings are mixed. Recently, we found that use of NSAIDs was differentially associated with lung cancer risk by histology. Here, we examine, more comprehensively, the association between individual NSAIDs and SCLC risk. 75,546 residents of western Washington State, ages 50-76, completed a baseline questionnaire in 2000-2002 and reported on their use of individual NSAIDs over the past 10 years. NSAID use was categorized as non-users, low (<4 days/week or <4 years), and high (≥4 days/week and ≥4 years). 111 SCLC were identified through linkage to a population-based cancer registry. Multivariable-adjusted Cox proportional hazards models including strong adjustment for smoking were used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI). Compared to non-use, high use of regular-strength aspirin was associated with an elevated risk of SCLC (HR 1.78, 95% CI: 1.05-3.02; P-trend=0.03). Findings for low-dose aspirin were elevated but did not reach statistical significance. Use of non-aspirin NSAIDs was not associated with SCLC risk. Our findings provide further indication of heterogeneity in the association between aspirin and lung cancer morphologies. Large, prospective studies with comprehensive assessments of NSAID use and smoking history and data on both men and women, are needed in order to better understand the association between use of aspirin and SCLC., (Copyright © 2012 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
35. Differentiation of human labia minora dermis-derived fibroblasts into insulin-producing cells.
- Author
-
Kim B, Yoon BS, Moon JH, Kim J, Jun EK, Lee JH, Kim JS, Baik CS, Kim A, Whang KY, and You S
- Subjects
- Animals, Biomarkers metabolism, Cell Proliferation drug effects, Cell Separation, Cells, Cultured, Dermis drug effects, Female, Fibroblasts drug effects, Glucose metabolism, Hepatocyte Nuclear Factor 3-beta metabolism, Homeodomain Proteins metabolism, Humans, Insulin metabolism, Insulin pharmacology, Insulin Secretion, Insulin-Secreting Cells metabolism, Mesenchymal Stem Cells drug effects, Mesenchymal Stem Cells metabolism, Mice, Mice, Nude, Niacinamide pharmacology, Recovery of Function, SOXF Transcription Factors metabolism, Sodium Selenite pharmacology, Trans-Activators metabolism, Transferrin pharmacology, Cell Culture Techniques, Cell Differentiation, Dermis cytology, Diabetes Mellitus, Experimental surgery, Fibroblasts cytology, Genitalia, Female cytology, Insulin-Secreting Cells cytology, Islets of Langerhans Transplantation, Mesenchymal Stem Cells cytology
- Abstract
Recent evidence has suggested that human skin fibroblasts may represent a novel source of therapeutic stem cells. In this study, we report a 3-stage method to induce the differentiation of skin fibroblasts into insulin- producing cells (IPCs). In stage 1, we establish the isolation, expansion and characterization of mesenchymal stem cells from human labia minora dermis- derived fibroblasts (hLMDFs) (stage 1: MSC expansion). hLMDFs express the typical mesenchymal stem cell marker proteins and can differentiate into adipocytes, osteoblasts, chondrocytes or muscle cells. In stage 2, DMEM/F12 serum-free medium with ITS mix (insulin, transferrin, and selenite) is used to induce differentiation of hLMDFs into endoderm-like cells, as determined by the expression of the endoderm markers Sox17, Foxa2, and PDX1 (stage 2: mesenchymal-endoderm transition). In stage 3, cells in the mesenchymal- endoderm transition stage are treated with nicotinamide in order to further differentiate into self-assembled, 3-dimensional islet cell-like clusters that express multiple genes related to pancreatic β-cell development and function (stage 3: IPC). We also found that the transplantation of IPCs can normalize blood glucose levels and rescue glucose homeostasis in streptozotocin- induced diabetic mice. These results indicate that hLMDFs have the capacity to differentiate into functionally competent IPCs and represent a potential cell-based treatment for diabetes mellitus.
- Published
- 2012
- Full Text
- View/download PDF
36. Reproductive factors, hormone use, and risk for lung cancer in postmenopausal women, the Nurses' Health Study.
- Author
-
Baik CS, Strauss GM, Speizer FE, and Feskanich D
- Subjects
- Adult, Age Factors, Aged, Aged, 80 and over, Estrogen Replacement Therapy adverse effects, Female, Humans, Middle Aged, Postmenopause, Risk Assessment, Risk Factors, Surveys and Questionnaires, United States epidemiology, Estrogen Replacement Therapy statistics & numerical data, Lung Neoplasms epidemiology, Reproductive History
- Abstract
Background: There is increasing evidence suggesting that female hormones may play a significant role in lung cancer development. We evaluated the associations between reproductive factors, exogenous hormone use, and lung cancer incidence in the Nurses' Health Study., Methods: We assessed age at menopause, age at menarche, type of menopause, parity, age at first birth, postmenopausal hormone (PMH) use, and past oral contraceptive use in 107,171 postmenopausal women. Cox models were used to estimate the hazard ratios for each exposure, adjusting for smoking and other covariates., Results: We identified 1,729 lung cancer cases during follow-up from 1984 to 2006. Menopause onset before 44 years of age (hazard ratio, 1.39; 95% confidence interval, 1.14-1.70) and past oral contraceptive use for >5 years (hazard ratio, 1.22; 95% confidence interval, 1.05-1.42) were associated with increased lung cancer risk. These associations were strongest in current smokers and small cell histology. In never smokers, increased parity was associated with decreased risk among parous women (P trend = 0.03), whereas in current smokers, older age at first birth was associated with increased risk (P trend = 0.02). PMH use was not associated with overall lung cancer incidence. However, nonsignificant results of increased risk in adenocarcinoma were seen with current PMH use., Conclusions: Our findings suggest female hormones may influence lung carcinogenesis, although the effect is likely modest, varied by histologic subtype, and altered by smoking., Impact: Further investigation of the pathophysiology of female hormones in lung cancer subtypes and their interaction with smoking will lead to better understanding of lung carcinogenesis., (©2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
37. Secretory profiles and wound healing effects of human amniotic fluid-derived mesenchymal stem cells.
- Author
-
Yoon BS, Moon JH, Jun EK, Kim J, Maeng I, Kim JS, Lee JH, Baik CS, Kim A, Cho KS, Lee JH, Lee HH, Whang KY, and You S
- Subjects
- Animals, Cell Differentiation drug effects, Cell Lineage drug effects, Cell Movement drug effects, Cell Proliferation drug effects, Cell Separation, Clone Cells, Culture Media, Conditioned pharmacology, Cytokines metabolism, Dermis cytology, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Humans, Mesenchymal Stem Cells drug effects, Mice, Mice, Inbred ICR, Protein Array Analysis, Smad2 Protein metabolism, Amniotic Fluid cytology, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Wound Healing drug effects
- Abstract
Recent evidence shows that amniotic fluid (AF) contains multiple cell types derived from the developing fetus, and may represent a novel source of stem cells for cell therapy. In this study, we examined the paracrine factors released by human amniotic fluid-derived mesenchymal stem cells (AF-MSCs) and their ability to accelerate the wound-healing process by stimulating proliferation and migration of dermal fibroblasts. AF-MSCs expressed the typical MSC marker proteins CD13, CD29, and CD44 and differentiated into adipocytes, osteoblasts, and chondrocytes when exposed to the appropriate differentiation media. In addition, AF-MSC-conditioned media (AF-MSC-CM) significantly enhanced proliferation of dermal fibroblasts. Antibody-based protein array and enzyme-linked immunosorbent assay (ELISA) indicated that AF-MSC-CM contains various cytokines and chemokines that are known to be important in normal wound healing, including IL-8, IL-6, TGF-beta, TNFRI, VEGF, and EGF. Application of AF-MSC-CM significantly enhanced wound healing by dermal fibroblasts via the TGF-beta/SMAD2 pathway. Levels of p-SMAD2 were increased by AF-MSC-CM, and both the increase in p-SMAD2 and migration of dermal fibroblasts were blocked by inhibiting the TGF-beta/SMAD2 pathway. Moreover, in a mouse excisional wound model, AF-MSC-CM accelerated wound healing. These data provide the first evidence of the potential for AF-MSC-CM in the treatment of skin wounds.
- Published
- 2010
- Full Text
- View/download PDF
38. [Down-Turner syndrome (45,X/47,XY,+21): case report and review].
- Author
-
Ryu SW, Lee G, Baik CS, Shim SH, Kim JT, Lee JS, and Lee KA
- Subjects
- Aneuploidy, Child, Preschool, Chromosome Banding, Chromosomes, Human, Pair 21, Chromosomes, Human, X, Chromosomes, Human, Y, Down Syndrome complications, Humans, In Situ Hybridization, Fluorescence, Karyotyping, Male, Trisomy, Turner Syndrome complications, Down Syndrome genetics, Mosaicism, Turner Syndrome genetics
- Abstract
We report the case of a 3-yr-old boy with Down-Turner mosaicism and review the previous reports of Down-Turner syndrome with documented karyotyping and clinical features. The patient showed clinical features of Down syndrome without significant stigma of Turner syndrome. Cytogenetic analysis of peripheral blood preparations by using G-banding revealed mosaicism with 2 cell lines (45,X[29]/47,XY,+21[4]). FISH analysis revealed that 87.5% of the cells had monosomy X karyotype and 12.5% of the cells had XY karyotype; trisomy 21 was only detected in the Y-positive cells. We suggest that additional cells should be analyzed and molecular genetic studies should be conducted to rule out double aneuploidy when karyotypes with sex chromosome aneuploidies and mosaicism are encountered, as in our case of Down syndrome mosaic with sex chromosome aneuploidy.
- Published
- 2010
- Full Text
- View/download PDF
39. Selection of optimal passage of bone marrow-derived mesenchymal stem cells for stem cell therapy in patients with amyotrophic lateral sclerosis.
- Author
-
Choi MR, Kim HY, Park JY, Lee TY, Baik CS, Chai YG, Jung KH, Park KS, Roh W, Kim KS, and Kim SH
- Subjects
- Cell Culture Techniques, Cell Differentiation, Cell Proliferation, Humans, Mesenchymal Stem Cell Transplantation, Time Factors, Amyotrophic Lateral Sclerosis therapy, Bone Marrow Cells cytology, Mesenchymal Stem Cells cytology
- Abstract
Mesenchymal stem cells (MSCs) obtained from bone marrow (BM) are currently used as an alternative therapy in amyotrophic lateral sclerosis (ALS) patients. Selection of optimal passages of autologous BM-derived MSCs during long-term in vitro expansion is important for clinical trials in patients with ALS. We isolated and expanded MSCs from the BM of eight ALS patients to analyze the growth kinetics, differentiation potential, cellular surface antigen expression, karyotype modifications and secretion of various cytokines during long-term culture. The morphology and size of the cells changed from small and spindle-like cells to large and polygonal types in later passages. The growth rate of the MSCs was highest in the third passage, followed by a gradual decrease. There were no special modifications of cell surface antigens or the karyotype of the MSCs from the first to the tenth passage. MSCs in the fourth passage were differentiated into adipocytes, osteocytes and chondrocytes. When we analyzed the cultured media of MSCs at the third, fifth, seventh and ninth passages, IL-6, VEGF and IL-8 showed high expression, with more than 50pg/10,000 cells at these passages; however, their expression progressively decreased with additional passages. In addition, secretion of IL-15, GM-CSF, IL-10, PDGF-bb, G-CSF, IL-1beta, basic FGF and IFN-gamma gradually decreased over prolonged culture. We suggest that MSCs at earlier passages are more suitable for stem cell therapy in ALS patients because of their stability and more potent anti-inflammatory and neuroprotective properties., (Copyright 2010 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
40. Effect of cytochalasin B and cycloheximide on the activation rate, chromosome constituent and in vitro development of porcine oocytes following parthenogenetic stimulation.
- Author
-
Cha SK, Kim NH, Lee SM, Baik CS, Lee HT, and Chung KS
- Subjects
- Animals, Cells, Cultured, Electric Stimulation, Female, Stimulation, Chemical, Swine, Chromosomes drug effects, Cycloheximide pharmacology, Cytochalasin B pharmacology, Oocytes drug effects, Parthenogenesis drug effects
- Abstract
Activation rate, chromosome constituent and developmental pattern of porcine oocytes was examined in the presence and absence of cytochalasin B and cycloheximide following parthenogenetic stimulation. Treatment with cycloheximide after ethanol or Ca2+ ionophore treatment increased the incidence of activation. The percentage of oocytes with two or more female pronuclei was higher (P < 0.05) in oocytes treated with cytochalasin B than in control or cycloheximide-treated oocytes. Treatment with both electrical stimulation and cytochalasin B increased the incidence of diploid chromosome spreads, and accelerated development to the morula and blastocyst stage compared with the control and cycloheximide-treated groups, suggesting a role of ploidy in the development of parthenote.
- Published
- 1997
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.