1
|
Lipsyc-Sharf M, Zhang S, Ou FS, Ma C, McCleary NJ, Niedzwiecki D, Chang IW, Lenz HJ, Blanke CD, Piawah S, Van Loon K, Bainter TM, Venook AP, Mayer RJ, Fuchs CS, Innocenti F, Nixon AB, Goldberg R, O’Reilly EM, Meyerhardt JA, Ng K. Survival in Young-Onset Metastatic Colorectal Cancer: Findings From Cancer and Leukemia Group B (Alliance)/SWOG 80405. J Natl Cancer Inst 2022; 114:427-435. [PMID: 34636852 PMCID: PMC8902338 DOI: 10.1093/jnci/djab200] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The incidence of young-onset colorectal cancer (yoCRC) is increasing. It is unknown if there are survival differences between young and older patients with metastatic colorectal cancer (mCRC). METHODS We studied the association of age with survival in 2326 mCRC patients enrolled in the Cancer and Leukemia Group B and SWOG 80405 trial, a multicenter, randomized trial of first-line chemotherapy plus biologics. The primary and secondary outcomes of this study were overall survival (OS) and progression-free survival (PFS), respectively, which were assessed by Kaplan-Meier method and compared among younger vs older patients with the log-rank test. Hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated based on Cox proportional hazards modeling, adjusting for known prognostic variables. All statistical tests were 2-sided. RESULTS Of 2326 eligible subjects, 514 (22.1%) were younger than age 50 years at study entry (yoCRC cohort). The median age of yoCRC patients was 44.3 vs 62.5 years in patients aged 50 years and older. There was no statistically significant difference in OS between yoCRC vs older-onset patients (median = 27.07 vs 26.12 months; adjusted HR = 0.98, 95% CI = 0.88 to 1.10; P = .78). The median PFS was also similar in yoCRC vs older patients (10.87 vs 10.55 months) with an adjusted hazard ratio of 1.02 (95% CI = 0.92 to 1.13; P = .67). Patients younger than age 35 years had the shortest OS with median OS of 21.95 vs 26.12 months in older-onset patients with an adjusted hazard ratio of 1.08 (95% CI = 0.81 to 1.44; Ptrend = .93). CONCLUSION In this large study of mCRC patients, there were no statistically significant differences in survival between patients with yoCRC and CRC patients aged 50 years and older.
Collapse
Affiliation(s)
- Marla Lipsyc-Sharf
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sui Zhang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Chao Ma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - I-Wen Chang
- Southeast Clinical Oncology Research (SCOR) Consortium, Winston-Salem, NC, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Charles D Blanke
- SWOG Group Chair’s Office/Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sorbarikor Piawah
- Department of Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Katherine Van Loon
- Department of Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Tiffany M Bainter
- Alliance Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - Alan P Venook
- Department of Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Charles S Fuchs
- Yale Cancer Center and Smilow Cancer Hospital, New Haven, CT, USA
- Genentech, South San Francisco, CA, USA
| | - Federico Innocenti
- Eshelman School of Pharmacy and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Eileen M O’Reilly
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA
| | | | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
2
|
Harbeck N, von Schumann R, Kates RE, Braun M, Kuemmel S, Schumacher C, Potenberg J, Malter W, Augustin D, Aktas B, Forstbauer H, Tio J, Grischke EM, Biehl C, Liedtke C, De Haas SL, Deurloo R, Wuerstlein R, Kreipe HH, Gluz O. Immune Markers and Tumor-Related Processes Predict Neoadjuvant Therapy Response in the WSG-ADAPT HER2-Positive/Hormone Receptor-Positive Trial in Early Breast Cancer. Cancers (Basel) 2021; 13:4884. [PMID: 34638369 PMCID: PMC8508505 DOI: 10.3390/cancers13194884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
Prognostic or predictive biomarkers in HER2-positive early breast cancer (EBC) may inform treatment optimization. The ADAPT HER2-positive/hormone receptor-positive phase II trial (NCT01779206) demonstrated pathological complete response (pCR) rates of ~40% following de-escalated treatment with 12 weeks neoadjuvant ado-trastuzumab emtansine (T-DM1) ± endocrine therapy. In this exploratory analysis, we evaluated potential early predictors of response to neoadjuvant therapy. The effects of PIK3CA mutations and immune (CD8 and PD-L1) and apoptotic markers (BCL2 and MCL1) on pCR rates were assessed, along with intrinsic BC subtypes. Immune response and pCR were lower in PIK3CA-mutated tumors compared with wildtype. Increased BCL2 at baseline in all patients and at Cycle 2 in the T-DM1 arms was associated with lower pCR. In the T-DM1 arms only, the HER2-enriched subtype was associated with increased pCR rate (54% vs. 28%). These findings support further prospective pCR-driven de-escalation studies in patients with HER2-positive EBC.
Collapse
Affiliation(s)
- Nadia Harbeck
- Breast Center, Department of Obstetrics and Gynecology and CCCLMU, University of Munich (LMU), Marchioninistrasse 15, 81377 Munich, Germany;
- The West German Study Group, 41061 Mönchengladbach, Germany; (R.E.K.); (O.G.)
| | - Raquel von Schumann
- Evangelical Hospital Bethesda, 41061 Mönchengladbach, Germany; (R.v.S.); (S.K.)
| | - Ronald Ernest Kates
- The West German Study Group, 41061 Mönchengladbach, Germany; (R.E.K.); (O.G.)
| | | | - Sherko Kuemmel
- Evangelical Hospital Bethesda, 41061 Mönchengladbach, Germany; (R.v.S.); (S.K.)
- Breast Unit, Kliniken Essen-Mitte, 45136 Essen, Germany
- Klinik für Gynäkologie mit Brustzentrum Charité-Universitätsmedizin, 10117 Berlin, Germany
| | | | | | - Wolfram Malter
- Breast Center, Department of Obstetrics and Gynecology, University Hospital Cologne, 50937 Cologne, Germany;
| | - Doris Augustin
- Breast Center, Clinic Deggendorf, 94469 Deggendorf, Germany;
| | | | | | - Joke Tio
- University Hospital Münster, 48149 Münster, Germany;
| | | | - Claudia Biehl
- Westphalian Breast Center, City Hospital Dortmund, 44137 Dortmund, Germany;
| | | | | | - Regula Deurloo
- F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland; (S.L.D.H.); (R.D.)
| | - Rachel Wuerstlein
- Breast Center, Department of Obstetrics and Gynecology and CCCLMU, University of Munich (LMU), Marchioninistrasse 15, 81377 Munich, Germany;
| | | | - Oleg Gluz
- The West German Study Group, 41061 Mönchengladbach, Germany; (R.E.K.); (O.G.)
- Evangelical Hospital Bethesda, 41061 Mönchengladbach, Germany; (R.v.S.); (S.K.)
- Breast Center, Department of Obstetrics and Gynecology, University Hospital Cologne, 50937 Cologne, Germany;
| |
Collapse
|
3
|
Guercio BJ, Zhang S, Venook AP, Ou FS, Niedzwiecki D, Lenz HJ, Innocenti F, Mullen BC, O'Neil BH, Shaw JE, Polite BN, Hochster HS, Atkins JN, Goldberg RM, Brown JC, O'Reilly EM, Mayer RJ, Blanke CD, Fuchs CS, Meyerhardt JA. Body Mass Index and Weight Loss in Metastatic Colorectal Cancer in CALGB (Alliance)/SWOG 80405. JNCI Cancer Spectr 2020; 4:pkaa024. [PMID: 33134818 PMCID: PMC7590517 DOI: 10.1093/jncics/pkaa024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/01/2020] [Accepted: 03/25/2020] [Indexed: 12/12/2022] Open
Abstract
Background In nonmetastatic colorectal cancer, overweight and mild-to-moderately obese patients experience improved outcomes compared with other patients. Obesity’s influence on advanced or metastatic colorectal cancer (mCRC) is relatively unexplored. Methods We conducted a prospective body mass index (BMI) companion study in Cancer and Leukemia Group B (now Alliance)/SWOG 80405, a phase III metastatic colorectal cancer (mCRC) treatment trial. BMI was measured at trial registration. Primary and secondary endpoints were overall and progression-free survival, respectively. To minimize confounding by poor and rapidly declining health, we used Cox proportional hazards regression to adjust for known prognostic factors, comorbidities, physical activity, and weight loss during the 6 months prior to study entry. We also examined weight loss prior to enrollment as an independent predictor of patient outcome. All statistical tests were two-sided. Results Among 2323 patients with mCRC, there were no statistically significant associations between BMI and overall or progression-free survival (adjusted Ptrend = .12 and .40, respectively). Weight loss during the 6 months prior to study entry was associated with shorter overall and progression-free survival; compared with individuals with stable weight ±4.9%, individuals with weight loss greater than 15% experienced an adjusted hazard ratio of 1.52 for all-cause mortality (95% confidence interval [CI] = 1.26 to 1.84; Ptrend < .001) and of 1.23 for disease progression or death (95% CI = 1.02 to 1.47; Ptrend = .006). Conclusions In this prospective study of patients with mCRC, BMI at time of first-line chemotherapy initiation was not associated with patient outcome. Weight loss prior to study entry was associated with increased risk of patient mortality and disease progression.
Collapse
Affiliation(s)
- Brendan J Guercio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Alan P Venook
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Rochester, MN, USA
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center, Duke University, Durham, NC, USA
| | | | | | - Brian C Mullen
- Alliance Statistics and Data Management Center, Rochester, MN, USA
| | - Bert H O'Neil
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - James E Shaw
- Medstar Washington Hospital Center, Washington, DC, USA
| | - Blase N Polite
- Pritzker School of Medicine, University of Chicago Comprehensive Cancer Center, Chicago, IL USA
| | | | - James N Atkins
- Southeast Clinical Oncology Research Consortium, NCORP, Winston-Salem, NC, USA
| | | | - Justin C Brown
- Department of Population and Public Health Science, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| | - Charles D Blanke
- SWOG Cancer Research Network and Oregon Health & Science University, Portland, OR, USA
| | - Charles S Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber/Partners CancerCare, Boston, MA, USA
| |
Collapse
|
4
|
Guercio BJ, Zhang S, Ou FS, Venook AP, Niedzwiecki D, Lenz HJ, Innocenti F, O'Neil BH, Shaw JE, Polite BN, Hochster HS, Atkins JN, Goldberg RM, Sato K, Ng K, Van Blarigan E, Mayer RJ, Blanke CD, O'Reilly EM, Fuchs CS, Meyerhardt JA. Associations of Physical Activity With Survival and Progression in Metastatic Colorectal Cancer: Results From Cancer and Leukemia Group B (Alliance)/SWOG 80405. J Clin Oncol 2019; 37:2620-2631. [PMID: 31408415 DOI: 10.1200/jco.19.01019] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Regular physical activity is associated with reduced risk of recurrence and mortality in patients with nonmetastatic colorectal cancer. Its influence on patients with advanced/metastatic colorectal cancer (mCRC) has been largely unexplored. PATIENTS AND METHODS We conducted a prospective cohort study nested in Cancer and Leukemia Group B (Alliance)/SWOG 80405 (ClinicalTrials.gov identifier: NCT00265850), a National Cancer Institute-sponsored phase III trial of systemic therapy for mCRC. Within 1 month after therapy initiation, patients were invited to complete a validated questionnaire that reported average physical activity over the previous 2 months. On the basis of responses, we calculated metabolic equivalent task (MET) hours per week to quantify physical activity. The primary end point of the clinical trial and this companion study was overall survival (OS). Secondary end points included progression-free survival (PFS) and first grade 3 or greater treatment-related adverse events. To minimize confounding by poor and declining health, we excluded patients who experienced progression or died within 60 days of activity assessment and used Cox proportional hazards regression analysis to adjust for known prognostic factors, comorbidities, and weight loss. RESULTS The final cohort included 1,218 patients. Compared with patients engaged in less than 3 MET hours per week of physical activity, patients engaged in 18 or more MET hours per week experienced an adjusted hazard ratio for OS of 0.85 (95% CI, 0.71 to 1.02; PTrend = .06) and for PFS of 0.83 (95% CI, 0.70 to 0.99; PTrend = .01). Compared with patients engaging in less than 9 MET hours per week, patients engaging in 9 or more MET hours per week experienced an adjusted hazard ratio for grade 3 or greater treatment-related adverse events of 0.73 (95% CI, 0.62 to 0.86; PTrend < .001). CONCLUSION Among patients with mCRC in Cancer and Leukemia Group B (Alliance)/SWOG 80405, association of physical activity with OS was not statistically significant. Greater physical activity was associated with longer PFS and lower adjusted risk for first grade 3 or greater treatment-related adverse events.
Collapse
Affiliation(s)
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | - Alan P Venook
- University of California, San Francisco, San Francisco, CA
| | | | - Heinz-Josef Lenz
- University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA
| | | | - Bert H O'Neil
- Indiana University School of Medicine, Indianapolis, IN
| | - James E Shaw
- Medstar Washington Hospital Center, Washington, DC
| | | | | | - James N Atkins
- Southeast Clinical Oncology Research Consortium, National Cancer Institute Community Oncology Research Program, Winston-Salem, NC
| | | | - Kaori Sato
- Dana-Farber/Partners CancerCare, Boston, MA
| | - Kimmie Ng
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | | | - Charles D Blanke
- SWOG, Portland, OR.,Oregon Health & Science University, Portland, OR
| | | | | | | |
Collapse
|
5
|
Theelen WSME, Kuilman T, Schulze K, Zou W, Krijgsman O, Peters DDGC, Cornelissen S, Monkhorst K, Sarma P, Sumiyoshi T, Amler LC, Willems SM, Blaauwgeers JLG, van Noesel CJM, Peeper DS, van den Heuvel MM, Kowanetz M. Absence of PD-L1 expression on tumor cells in the context of an activated immune infiltrate may indicate impaired IFNγ signaling in non-small cell lung cancer. PLoS One 2019; 14:e0216864. [PMID: 31125352 PMCID: PMC6534376 DOI: 10.1371/journal.pone.0216864] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 04/30/2019] [Indexed: 11/18/2022] Open
Abstract
Background In non-small cell lung cancer (NSCLC), PD-L1 expression on either tumor cells (TC) or both TC and tumor-infiltrating immune cells (IC) is currently the most used biomarker in cancer immunotherapy. However, the mechanisms involved in PD-L1 regulation are not fully understood. To provide better insight in these mechanisms, a multiangular analysis approach was used to combine protein and mRNA expression with several clinicopathological characteristics. Patients and methods Archival tissues from 640 early stage, resected NSCLC patients were analyzed with immunohistochemistry for expression of PD-L1 and CD8 infiltration. In addition, mutational status and expression of a selection of immune genes involved in the PD-L1/PD-1 axis and T-cell response was determined. Results Tumors with high PD-L1 expression on TC or on IC represent two subsets of NSCLC with minimal overlap. We observed that PD-L1 expression on IC irrespective of expression on TC is a good marker for inflammation within tumors. In the tumors with the highest IC expression and absent TC expression an association with reduced IFNγ downstream signaling in tumor cells was observed. Conclusions These results show that PD-L1 expression on TC and IC are both independent hallmarks of the inflamed phenotype in NSCLC, and TC-negative/IC-high tumors can also be categorized as inflamed. The lack of correlation between PD-L1 TC and IC expression in this subgroup may be caused by impaired IFNγ signaling in tumor cells. These findings may bring a better understanding of the tumor-immune system interaction and the clinical relevance of PD-L1 expression on IC irrespective of PD-L1 expression on TC.
Collapse
Affiliation(s)
| | - Thomas Kuilman
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Katja Schulze
- Oncology Biomarker Development, Genentech Inc., South San Francisco, United States of America
| | - Wei Zou
- Biostatistics, Genentech Inc., South San Francisco, United States of America
| | - Oscar Krijgsman
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Dennis D. G. C. Peters
- Core Facility Molecular Pathology & Biobanking, Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Sten Cornelissen
- Core Facility Molecular Pathology & Biobanking, Department of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Kim Monkhorst
- Division of Pathology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Pranamee Sarma
- Oncology Biomarker Development, Genentech Inc., South San Francisco, United States of America
| | - Teiko Sumiyoshi
- Oncology Biomarker Development, Genentech Inc., South San Francisco, United States of America
| | - Lukas C. Amler
- Oncology Biomarker Development, Genentech Inc., South San Francisco, United States of America
| | - Stefan M. Willems
- Department of Pathology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | | | | | - Daniel S. Peeper
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Marcin Kowanetz
- Oncology Biomarker Development, Genentech Inc., South San Francisco, United States of America
| |
Collapse
|
6
|
Piskol R, Huw L, Sergin I, Kljin C, Modrusan Z, Kim D, Kljavin N, Tam R, Patel R, Burton J, Penuel E, Qu X, Koeppen H, Sumiyoshi T, de Sauvage F, Lackner MR, de Sousa e Melo F, Kabbarah O. A Clinically Applicable Gene-Expression Classifier Reveals Intrinsic and Extrinsic Contributions to Consensus Molecular Subtypes in Primary and Metastatic Colon Cancer. Clin Cancer Res 2019; 25:4431-4442. [DOI: 10.1158/1078-0432.ccr-18-3032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/29/2019] [Accepted: 04/15/2019] [Indexed: 01/10/2023]
|
7
|
Innocenti F, Ou FS, Qu X, Zemla TJ, Niedzwiecki D, Tam R, Mahajan S, Goldberg RM, Bertagnolli MM, Blanke CD, Sanoff H, Atkins J, Polite B, Venook AP, Lenz HJ, Kabbarah O. Mutational Analysis of Patients With Colorectal Cancer in CALGB/SWOG 80405 Identifies New Roles of Microsatellite Instability and Tumor Mutational Burden for Patient Outcome. J Clin Oncol 2019; 37:1217-1227. [PMID: 30865548 DOI: 10.1200/jco.18.01798] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE CALGB/SWOG 80405 was a randomized phase III trial that found no statistically significant difference in overall survival (OS) in patients with first-line metastatic colorectal cancer treated with chemotherapy plus either bevacizumab or cetuximab. Primary tumor DNA from 843 patients has been used to discover genetic markers of OS. PATIENTS AND METHODS Gene mutations were determined by polymerase chain reaction. Microsatellite status was determined by genotyping of microsatellites. Tumor mutational burden (TMB) was determined by next-generation sequencing. Cox proportional hazard models were used, with adjusting factors. Interaction of molecular alterations with either the bevacizumab or the cetuximab arms was tested. RESULTS Patients with high TMB in their tumors had longer OS than did patients with low TMB (hazard ratio [HR], 0.73 [95% CI, 0.57 to 0.95]; P = .02). In patients with microsatellite instability-high (MSI-H) tumors, longer OS was observed in the bevacizumab arm than in the cetuximab arm (HR, 0.13 [95% CI, 0.06 to 0.30]; interaction P < .001 for interaction between microsatellite status and the two arms). Patients with BRAF mutant tumors had shorter OS than did patients with wild-type (WT) tumors (HR, 2.01 [95% CI, 1.49 to 2.71]; P < .001). Patients with extended RAS mutant tumors had shorter OS than did patients with WT tumors (HR, 1.52 [95% CI, 1.26 to 1.84]; P < .001). Patients with triple-negative tumors (WT for NRAS/KRAS/BRAF) had a median OS of 35.9 months (95% CI, 33.0 to 38.8 months) versus 22.2 months (95% CI, 19.6 to 24.4 months ) in patients with at least one mutated gene in their tumors (P < .001). CONCLUSION In patients with metastatic colorectal cancer treated in first line, low TMB, and BRAF and RAS mutations are negative prognostic factors. Patients with MSI-H tumors benefited more from bevacizumab than from cetuximab, and studies to confirm this effect of MSI-H are warranted.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hanna Sanoff
- 1 University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - James Atkins
- 8 Southeast Clinical Oncology Research Consortium, Winston-Salem, NC
| | - Blasé Polite
- 9 University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Alan P Venook
- 10 University of California San Francisco, San Francisco, CA
| | | | | |
Collapse
|
8
|
Phase II Study of the Dual EGFR/HER3 Inhibitor Duligotuzumab (MEHD7945A) versus Cetuximab in Combination with FOLFIRI in Second-Line RAS Wild-Type Metastatic Colorectal Cancer. Clin Cancer Res 2018; 24:2276-2284. [DOI: 10.1158/1078-0432.ccr-17-0646] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 10/31/2017] [Accepted: 02/26/2018] [Indexed: 11/16/2022]
|
9
|
Yamasaki M, Funaishi K, Saito N, Sakano A, Fujihara M, Daido W, Ishiyama S, Deguchi N, Taniwaki M, Ohashi N, Hattori N. Putative lung adenocarcinoma with epidermal growth factor receptor mutation presenting as carcinoma of unknown primary site: A case report. Medicine (Baltimore) 2018; 97:e9942. [PMID: 29443782 PMCID: PMC5839817 DOI: 10.1097/md.0000000000009942] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
RATIONALE Only a few cases of putative lung adenocarcinoma presenting as carcinoma of unknown primary site (CUP) with epidermal growth factor receptor (EGFR) mutation have been reported, and the efficacy of EGFR-tyrosine kinase inhibitors (TKIs) for these cases is unclear. PATIENT CONCERNS AND DIAGNOSES A 67-year-old man complained of paresis of the right lower extremity, dysarthria, and memory disturbance. Computed tomography and magnetic resonance imaging showed multiple brain tumors with brain edema and swelling of the left supraclavicular, mediastinal, and upper abdominal lymph nodes. Moreover, a metastatic duodenal tumor was detected via upper gastrointestinal endoscopy examination. The biopsy specimen of the lesion was examined and was diagnosed as adenocarcinoma with CK7 and TTF-1 positivity. Finally, the case was diagnosed as EGFR mutation-positive putative lung adenocarcinoma presenting as CUP. INTERVENTIONS AND OUTCOMES Oral erlotinib, an EGFR-TKI, was administered at 150 mg daily. Five weeks later, the brain lesions and several swollen lymph nodes showed marked improvement, and the symptoms of the patient also improved. Three months later, the duodenal lesion was undetected on upper gastrointestinal endoscopy. After an 8-month follow-up, the patient was well with no disease progression. LESSONS Putative lung adenocarcinoma presenting as CUP may have EGFR mutation, and EGFR-TKI therapy may be effective for such malignancy.
Collapse
Affiliation(s)
- Masahiro Yamasaki
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Kunihiko Funaishi
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Naomi Saito
- Department of Respiratory Medicine, Mazda Hospital, Aki-gun
| | | | - Megumu Fujihara
- Department of Pathology, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital
| | - Wakako Daido
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Sayaka Ishiyama
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Naoko Deguchi
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Masaya Taniwaki
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
| | - Nobuyuki Ohashi
- Department of Respiratory Disease, Hiroshima Red Cross Hospital & Atomic Bomb Survivors Hospital, Naka-ku
- Ohashi Clinic, Naka-ku
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Institute of Biomedical & Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| |
Collapse
|
10
|
Fumagalli D, Wilson TR, Salgado R, Lu X, Yu J, O'Brien C, Walter K, Huw LY, Criscitiello C, Laios I, Jose V, Brown DN, Rothé F, Maetens M, Zardavas D, Savas P, Larsimont D, Piccart-Gebhart MJ, Michiels S, Lackner MR, Sotiriou C, Loi S. Somatic mutation, copy number and transcriptomic profiles of primary and matched metastatic estrogen receptor-positive breast cancers. Ann Oncol 2017; 27:1860-6. [PMID: 27672107 DOI: 10.1093/annonc/mdw286] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 07/14/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Estrogen receptor-positive (ER+) breast cancers (BCs) constitute the most frequent BC subtype. The molecular landscape of ER+ relapsed disease is not well characterized. In this study, we aimed to describe the genomic evolution between primary (P) and matched metastatic (M) ER+ BCs after failure of adjuvant therapy. MATERIALS AND METHODS A total of 182 ER+ metastatic BC patients with long-term follow-up were identified from a single institution. P tumor tissue was available for all patients, with 88 having matched M material. According to the availability of tumor material, samples were characterized using a 120 mutational hotspot qPCR, a 29 gene copy number aberrations (CNA) and a 400 gene expression panels. ESR1 mutations were assayed by droplet digital PCR. Molecular alterations were correlated with overall survival (OS) using the Cox proportional hazards regression models. RESULTS The median follow-up was 6.4 years (range 0.5-26.6 years). Genomic analysis of P tumors revealed somatic mutations in PIK3CA, KRAS, AKT1, FGFR3, HRAS and BRAF at frequencies of 41%, 6%, 5%, 2%, 1% and 2%, respectively, and CN amplification of CCND1, ZNF703, FGFR1, RSF1 and PAK1 at 23%, 19%, 17%, 12% and 11%, respectively. Mutations and CN amplifications were largely concordant between P and matched M (>84%). ESR1 mutations were found in 10.8% of the M but none of the P. Thirteen genes, among which ESR1, FOXA1, and HIF1A, showed significant differential expression between P and M. In P, the differential expression of 18 genes, among which IDO1, was significantly associated with OS (FDR < 0.1). CONCLUSIONS Despite the large concordance between P and matched M for the evaluated molecular alterations, potential actionable targets such as ESR1 mutations were found only in M. This supports the importance of characterizing the M disease. Other targets we identified, such as HIF1A and IDO1, warrant further investigation in this patient population.
Collapse
Affiliation(s)
- D Fumagalli
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - T R Wilson
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - R Salgado
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - X Lu
- Department of Biostatistics, Genentech Inc., South San Francisco, CA, USA
| | - J Yu
- Department of Biostatistics, Genentech Inc., South San Francisco, CA, USA
| | - C O'Brien
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - K Walter
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - L Y Huw
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - C Criscitiello
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, Milan, Italy
| | - I Laios
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | - V Jose
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - D N Brown
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - F Rothé
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - M Maetens
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium
| | - D Zardavas
- Breast International Group, Brussels, Belgium
| | - P Savas
- Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | - D Larsimont
- Department of Pathology, Institut Jules Bordet, Brussels, Belgium
| | | | - S Michiels
- Division of Biostatistics and Epidemiology, Institut Gustave Roussy, Villejuif, France INSERM U1018, CESP, University of Paris, Villejuif, France
| | - M R Lackner
- Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - C Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Free University of Bruxelles, Brussels, Belgium Division of Medical Oncology, Institut Jules Bordet, Brussels, Belgium
| | - S Loi
- Division of Clinical Medicine and Research, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Juric D, Krop I, Ramanathan RK, Wilson TR, Ware JA, Sanabria Bohorquez SM, Savage HM, Sampath D, Salphati L, Lin RS, Jin H, Parmar H, Hsu JY, Von Hoff DD, Baselga J. Phase I Dose-Escalation Study of Taselisib, an Oral PI3K Inhibitor, in Patients with Advanced Solid Tumors. Cancer Discov 2017; 7:704-715. [PMID: 28331003 DOI: 10.1158/2159-8290.cd-16-1080] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 12/06/2016] [Accepted: 03/20/2017] [Indexed: 12/31/2022]
Abstract
Taselisib is a potent and selective tumor growth inhibitor through PI3K pathway suppression. Thirty-four patients with locally advanced or metastatic solid tumors were treated (phase I study, modified 3+3 dose escalation; 5 cohorts; 3-16 mg taselisib once-daily capsule). Taselisib pharmacokinetics were dose-proportional; mean half-life was 40 hours. Frequent dose-dependent, treatment-related adverse events included diarrhea, hyperglycemia, decreased appetite, nausea, rash, stomatitis, and vomiting. At 12 and 16 mg dose levels, dose-limiting toxicities (DLT) were observed, with an accumulation of higher-grade adverse events after the cycle 1 DLT assessment window. Pharmacodynamic findings showed pathway inhibition at ≥3 mg in patient tumor samples, consistent with preclinical PIK3CA-mutant tumor xenograft models. Confirmed response rate was 36% for PIK3CA-mutant tumor patients with measurable disease [5/14: 4 breast cancer (3 patients at 12 mg); 1 non-small cell lung cancer], where responses started at 3 mg, and 0% in patients with tumors without known PIK3CA hotspot mutations (0/15).Significance: Preliminary data consistent with preclinical data indicate increased antitumor activity of taselisib in patients with PIK3CA-mutant tumors (in comparison with patients with tumors without known activating PIK3CA hotspot mutations) starting at the lowest dose tested of 3 mg, thereby supporting higher potency for taselisib against PIK3CA-mutant tumors. Cancer Discov; 7(7); 704-15. ©2017 AACR.See related commentary by Rodon and Tabernero, p. 666This article is highlighted in the In This Issue feature, p. 653.
Collapse
Affiliation(s)
- Dejan Juric
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ian Krop
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | | | | | | | - Ray S Lin
- Genentech, Inc., South San Francisco, California
| | - Huan Jin
- Genentech, Inc., South San Francisco, California
| | - Hema Parmar
- Genentech, Inc., South San Francisco, California
| | - Jerry Y Hsu
- Genentech, Inc., South San Francisco, California
| | | | - José Baselga
- Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
12
|
Kim D, Choi Y, Ireland J, Foreman O, Tam RN, Patel R, Schleifman EB, Motlhabi M, French D, Wong CV, Peters E, Molinero L, Raja R, Amler LC, Hampton GM, Lackner MR, Kabbarah O. Development and Application of a Microfluidics-Based Panel in the Basal/Luminal Transcriptional Characterization of Archival Bladder Cancers. PLoS One 2016; 11:e0165856. [PMID: 27846280 PMCID: PMC5112874 DOI: 10.1371/journal.pone.0165856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 10/19/2016] [Indexed: 11/18/2022] Open
Abstract
In the age of personalized medicine stratifying tumors into molecularly defined subtypes associated with distinctive clinical behaviors and predictable responses to therapies holds tremendous value. Towards this end, we developed a custom microfluidics-based bladder cancer gene expression panel for characterization of archival clinical samples. In silico analysis indicated that the content of our panel was capable of accurately segregating bladder cancers from several public datasets into the clinically relevant basal and luminal subtypes. On a technical level, our bladder cancer panel yielded robust and reproducible results when analyzing formalin-fixed, paraffin-embedded (FFPE) tissues. We applied our panel in the analysis of a novel set of 204 FFPE samples that included non-muscle invasive bladder cancers (NMIBCs), muscle invasive disease (MIBCs), and bladder cancer metastases (METs). We found NMIBCs to be mostly luminal-like, MIBCs to include both luminal- and basal-like types, and METs to be predominantly of a basal-like transcriptional profile. Mutational analysis confirmed the expected enrichment of FGFR3 mutations in luminal samples, and, consistently, FGFR3 IHC showed high protein expression levels of the receptor in these tumors. Our bladder cancer panel enables basal/luminal characterization of FFPE tissues and with further development could be used for stratification of bladder cancer samples in the clinic.
Collapse
Affiliation(s)
- Doris Kim
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - YounJeong Choi
- Department of Biostatistics, Genentech, South San Francisco, California, United States of America
| | - James Ireland
- Alternate Allele Consulting, Orinda, California, United States of America
| | - Oded Foreman
- Department of Pathology, Genentech, South San Francisco, California, United States of America
| | - Rachel N Tam
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Rajesh Patel
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Erica B Schleifman
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Maipelo Motlhabi
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Dorothy French
- Department of Pathology, Genentech, South San Francisco, California, United States of America
| | - Cheryl V Wong
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Eric Peters
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Luciana Molinero
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Rajiv Raja
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Lukas C Amler
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Garret M Hampton
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Mark R Lackner
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| | - Omar Kabbarah
- Oncology Biomarker Development, Genentech, South San Francisco, California, United States of America
| |
Collapse
|
13
|
The molecular landscape of high-risk early breast cancer: comprehensive biomarker analysis of a phase III adjuvant population. NPJ Breast Cancer 2016; 2:16022. [PMID: 28721382 PMCID: PMC5515335 DOI: 10.1038/npjbcancer.2016.22] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 06/03/2016] [Accepted: 06/10/2016] [Indexed: 01/22/2023] Open
Abstract
Breast cancer is a heterogeneous disease and patients are managed clinically based on ER, PR, HER2 expression, and key risk factors. We sought to characterize the molecular landscape of high-risk breast cancer patients enrolled onto an adjuvant chemotherapy study to understand how disease subsets and tumor immune status impact survival. DNA and RNA were extracted from 861 breast cancer samples from patients enrolled onto the United States Oncology trial 01062. Samples were characterized using multiplex gene expression, copy number, and qPCR mutation assays. HR+ patients with a PIK3CA mutant tumor had a favorable disease-free survival (DFS; HR 0.66, P=0.05), however, the prognostic effect was specific to luminal A patients (Luminal A: HR 0.67, P=0.1; Luminal B: HR 1.01, P=0.98). Molecular subtyping of triple-negative breast cancers (TNBCs) suggested that the mesenchymal subtype had the worst DFS, whereas the immunomodulatory subtype had the best DFS. Profiling of immunologic genes revealed that TNBC tumors (n=280) displaying an activated T-cell signature had a longer DFS following adjuvant chemotherapy (HR 0.59, P=0.04), while a distinct set of immune genes was associated with DFS in HR+ cancers. Utilizing a discovery approach, we identified genes associated with a high risk of recurrence in HR+ patients, which were validated in an independent data set. Molecular classification based on PAM50 and TNBC subtyping stratified clinical high-risk patients into distinct prognostic subsets. Patients with high expression of immune-related genes showed superior DFS in both HR+ and TNBC. These results may inform patient management and drug development in early breast cancer.
Collapse
|
14
|
Houghton J, Hadd AG, Zeigler R, Haynes BC, Latham GJ. Integration of Wet and Dry Bench Processes Optimizes Targeted Next-generation Sequencing of Low-quality and Low-quantity Tumor Biopsies. J Vis Exp 2016:e53836. [PMID: 27166994 PMCID: PMC4941914 DOI: 10.3791/53836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
All next-generation sequencing (NGS) procedures include assays performed at the laboratory bench ("wet bench") and data analyses conducted using bioinformatics pipelines ("dry bench"). Both elements are essential to produce accurate and reliable results, which are particularly critical for clinical laboratories. Targeted NGS technologies have increasingly found favor in oncology applications to help advance precision medicine objectives, yet the methods often involve disconnected and variable wet and dry bench workflows and uncoordinated reagent sets. In this report, we describe a method for sequencing challenging cancer specimens with a 21-gene panel as an example of a comprehensive targeted NGS system. The system integrates functional DNA quantification and qualification, single-tube multiplexed PCR enrichment, and library purification and normalization using analytically-verified, single-source reagents with a standalone bioinformatics suite. As a result, accurate variant calls from low-quality and low-quantity formalin-fixed, paraffin-embedded (FFPE) and fine-needle aspiration (FNA) tumor biopsies can be achieved. The method can routinely assess cancer-associated variants from an input of 400 amplifiable DNA copies, and is modular in design to accommodate new gene content. Two different types of analytically-defined controls provide quality assurance and help safeguard call accuracy with clinically-relevant samples. A flexible "tag" PCR step embeds platform-specific adaptors and index codes to allow sample barcoding and compatibility with common benchtop NGS instruments. Importantly, the protocol is streamlined and can produce 24 sequence-ready libraries in a single day. Finally, the approach links wet and dry bench processes by incorporating pre-analytical sample quality control results directly into the variant calling algorithms to improve mutation detection accuracy and differentiate false-negative and indeterminate calls. This targeted NGS method uses advances in both wetware and software to achieve high-depth, multiplexed sequencing and sensitive analysis of heterogeneous cancer samples for diagnostic applications.
Collapse
|
15
|
Raub CB, Lee CC, Shibata D, Taylor C, Kartalov E. HistoMosaic Detecting KRAS G12V Mutation Across Colorectal Cancer Tissue Slices through in Situ PCR. Anal Chem 2016; 88:2792-8. [PMID: 26820161 PMCID: PMC7446286 DOI: 10.1021/acs.analchem.5b04460] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
We report on HistoMosaic, a novel technique for genetic analysis of formalin-fixed, paraffin-embedded tissue slices. It combines microfluidic compartmentalization, in situ allele-specific PCR, and fluorescence microscopy. The experimental proof of principle was achieved by in situ detection of KRAS G12V mutation in colorectal cancer tissues and is presented herein. HistoMosaic offers the ability to detect mutations over the entire tissue slide simultaneously, rapidly, economically, and without selection bias, while coregistering the genetic information with the preserved morphological information. Thus, HistoMosaic has wide applicability in basic science as a tool to map genetic heterogeneity. It is also a platform to build companion diagnostics for targeted therapies in oncology, to help ensure that the right drug is given to the right patient, thereby saving healthcare resources and improving patient outcomes.
Collapse
Affiliation(s)
| | - Chen-Chung Lee
- Department of Pathology, Keck School of Medicine of the University of Southern California, 2011 Zonal Avenue, Los Angeles, California 90089, United States
| | - Darryl Shibata
- Department of Pathology, Keck School of Medicine of the University of Southern California, 2011 Zonal Avenue, Los Angeles, California 90089, United States
| | - Clive Taylor
- Department of Pathology, Keck School of Medicine of the University of Southern California, 2011 Zonal Avenue, Los Angeles, California 90089, United States
| | - Emil Kartalov
- Department of Pathology, Keck School of Medicine of the University of Southern California, 2011 Zonal Avenue, Los Angeles, California 90089, United States
| |
Collapse
|
16
|
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independentei, 050096, Bucharest, Romania
| | - Radu Albulescu
- Biochemistry-Proteomics Laboratory, “Victor Babes” National Institute of Pathology & National Institute for Chemical Pharmaceutical R&D Bucharest
| | - Cristiana Tanase
- Biochemistry-Proteomics Laboratory, “Victor Babes” National Institute of Pathology & “Titu Maiorescu” University, Faculty of Medicine
| |
Collapse
|
17
|
Kawano Y, Otsuka C, Sanzo J, Higgins C, Nirei T, Schilling T, Ishikawa T. Expanding imaging capabilities for microfluidics: applicability of darkfield internal reflection illumination (DIRI) to observations in microfluidics. PLoS One 2015; 10:e0116925. [PMID: 25748425 PMCID: PMC4352060 DOI: 10.1371/journal.pone.0116925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 12/16/2014] [Indexed: 01/09/2023] Open
Abstract
Microfluidics is used increasingly for engineering and biomedical applications due to recent advances in microfabrication technologies. Visualization of bubbles, tracer particles, and cells in a microfluidic device is important for designing a device and analyzing results. However, with conventional methods, it is difficult to observe the channel geometry and such particles simultaneously. To overcome this limitation, we developed a Darkfield Internal Reflection Illumination (DIRI) system that improved the drawbacks of a conventional darkfield illuminator. This study was performed to investigate its utility in the field of microfluidics. The results showed that the developed system could clearly visualize both microbubbles and the channel wall by utilizing brightfield and DIRI illumination simultaneously. The methodology is useful not only for static phenomena, such as clogging, but also for dynamic phenomena, such as the detection of bubbles flowing in a channel. The system was also applied to simultaneous fluorescence and DIRI imaging. Fluorescent tracer beads and channel walls were observed clearly, which may be an advantage for future microparticle image velocimetry (μPIV) analysis, especially near a wall. Two types of cell stained with different colors, and the channel wall, can be recognized using the combined confocal and DIRI system. Whole-slide imaging was also conducted successfully using this system. The tiling function significantly expands the observing area of microfluidics. The developed system will be useful for a wide variety of engineering and biomedical applications for the growing field of microfluidics.
Collapse
Affiliation(s)
- Yoshihiro Kawano
- The Department of Biomedical Engineering, Graduate School of Biomedical Engineering, Tohoku University, Sendai, Miyagi, Japan; Olympus Corporation, Shinjuku-Ku, Tokyo, Japan
| | | | - James Sanzo
- Olympus Scientific Solutions Americas, Waltham, Massachusetts, United States of America
| | - Christopher Higgins
- Olympus Scientific Solutions Americas, Waltham, Massachusetts, United States of America
| | | | | | - Takuji Ishikawa
- Department of Bioengineering and Robotics, Tohoku University, Sendai, Miyagi, Japan
| |
Collapse
|