199 results on '"Geoffrey Y. Ku"'
Search Results
2. Nanoliposomal irinotecan with fluorouracil for the treatment of advanced pancreatic cancer, a single institution experience
- Author
-
Danielle C. Glassman, Randze L. Palmaira, Christina M. Covington, Avni M. Desai, Geoffrey Y. Ku, Jia Li, James J. Harding, Anna M. Varghese, Eileen M. O’Reilly, and Kenneth H. Yu
- Subjects
Pancreatic cancer ,Nanoliposomal irinotecan ,MM-398 ,Nal-IRI ,5-fluorouracil ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Effective treatment options for advanced pancreatic cancer are finite. NAPOLI-1, a phase III randomized trial, demonstrated the efficacy of nanoliposomal irinotecan with fluorouracil/leucovorin (nal-IRI + 5-FU/LV) for the treatment of advanced pancreatic cancer following progression on gemcitabine-based chemotherapy. There are limited additional data on the safety and efficacy of nal-IRI + 5-FU/LV following FDA approval in October 2015. We examined the post-approval safety and effectiveness of nal-IRI + 5-FU/LV in advanced pancreatic cancer patients receiving treatment at Memorial Sloan Kettering Cancer Center. Methods A retrospective chart review was conducted of all patients beginning treatment with nal-IRI + 5-FU/LV from October 2015 through June 2017. Using the electronic medical record and institutional database, information was extracted pertaining to demographics, performance status (ECOG), prior therapies, dose, duration of treatment, adverse events, progression free survival (PFS), overall survival (OS) and treatment response. Results Fifty six patients were identified. Median progression free survival (PFS) was 2.9 months and median overall survival (OS) was 5.3 months. Patients with prior disease progression on irinotecan experienced PFS and OS of 2.2 and 3.9 mo, respectively. Patients without prior irinotecan exposure experienced significantly longer PFS (4.8 mo, p = 0.02) and OS (7.7 mo, p = 0.002), as did patients who received prior irinotecan without disease progression (PFS, 5.7 mo, p = 0.04; OS, 9.0 mo, p = .04). Progression on prior irinotecan was associated with greater lines of prior advanced disease chemotherapy (2 vs 1). Dose reductions (DR) were most frequently due to fatigue (42%) and diarrhea (37%), but were not associated with worse outcomes. In fact, patients with ≥1 DR experienced longer PFS (5.4 v 2.6 mo, p = 0.035). Sequential therapy with nab-paclitaxel + gemcitabine (nab-P + Gem) followed by nal-IRI + 5-FU/LV (n = 25) resulted in OS of 23.0 mo. Mutations in TP53 were associated with shorter PFS. Conclusions These data support the safety and efficacy of nal-IRI + 5-FU/LV, reinforcing results of NAPOLI-1. Patients without disease progression on prior irinotecan fared significantly better than patients with progression, when treated with nal-IRI + 5-FU/LV. Sequential therapy with nab-P + Gem followed by nal-IRI + 5-FU/LV demonstrates encouraging median OS. These findings provide guidance for patients most likely to benefit from nal-IRI + 5-FU/LV.
- Published
- 2018
- Full Text
- View/download PDF
3. Survival After Trimodality Therapy in Patients With Locally Advanced Esophagogastric Adenocarcinoma: Does Only a Complete Pathologic Response Matter?
- Author
-
Smita Sihag, Geoffrey Y. Ku, Kay See Tan, Abraham J. Wu, Yelena Y. Janjigian, Steven Brad Maron, David R. Jones, Laura H. Tang, Daniela Molena, Tamar B. Nobel, Manjit S. Bains, Sergio De La Torre, and Meier Hsu
- Subjects
medicine.medical_specialty ,Neoplasm, Residual ,Esophageal Neoplasms ,Locally advanced ,Adenocarcinoma ,Gastroenterology ,Article ,03 medical and health sciences ,0302 clinical medicine ,Major Pathologic Response ,Internal medicine ,Clinical endpoint ,Medicine ,Humans ,Stage (cooking) ,Survival analysis ,Retrospective Studies ,Neoplasm Staging ,business.industry ,Remission Induction ,medicine.disease ,Primary tumor ,Neoadjuvant Therapy ,Clinical trial ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,Surgery ,business - Abstract
OBJECTIVE: To evaluate whether pathologic complete response (pCR) exclusively defines major pathologic response to treatment with improved survival. SUMMARY BACKGROUND DATA: pCR following trimodality therapy for esophagogastric adenocarcinoma is infrequent but associated with improved prognosis. Yet most clinical trials and correlative studies designate pCR as the primary endpoint. METHODS: We analyzed our prospectively maintained database for patients who underwent trimodality therapy for locally advanced esophageal adenocarcinoma between 1995 and 2017. Overall survival (OS) was examined by percentage treatment response (TR) in the primary tumor bed and pathologic nodal stage (ypN0) using Kaplan-Meier plots. Optimal thresholds of TR for differentiating patients in terms of OS were investigated with descriptive plots using restricted cubic spline functions; associations were quantified using Cox multivariable analysis. RESULTS: Among 788 patients, median follow-up was 37.5 months (range, 0.4–210.6); median OS was 48.3 months (95% CI, 42.2–58.8). Absence of residual nodal disease was independently associated with improved survival (P
- Published
- 2023
4. Epidermal Growth Factor Receptor Inhibition in Epidermal Growth Factor Receptor-Amplified Gastroesophageal Cancer: Retrospective Global Experience
- Author
-
Steven B. Maron, Stephanie Moya, Federica Morano, Matthew J. Emmett, Joanne F. Chou, Shalom Sabwa, Henry Walch, Bryan Peterson, Alexa B. Schrock, Liangliang Zhang, Yelena Y. Janjigian, Sree Chalasani, Geoffrey Y. Ku, Umut Disel, Peter Enzinger, Nataliya Uboha, Shumei Kato, Takayuki Yoshino, Kohei Shitara, Yoshiaki Nakamura, Anwaar Saeed, Pashtoon M. Kasi, Joseph Chao, Jeeyun Lee, Marinela Capanu, Zev Wainberg, Russell Petty, Filippo Pietrantonio, Samuel J. Klempner, and Daniel V.T. Catenacci
- Subjects
ErbB Receptors ,Cancer Research ,Oncology ,Esophageal Neoplasms ,Stomach Neoplasms ,Antibodies, Bispecific ,Humans ,Adenocarcinoma ,In Situ Hybridization, Fluorescence ,Retrospective Studies - Abstract
PURPOSE Subset analyses from phase III evaluation of epidermal growth factor receptor inhibition (EGFRi) suggest improved outcomes in patients with EGFR-amplified gastroesophageal adenocarcinoma (GEA), but large-scale analyses are lacking. This multi-institutional analysis sought to determine the role of EGFRi in the largest cohort of patients with EGFR-amplified GEA to date. PATIENTS AND METHODS A total of 60 patients from 15 tertiary cancer centers in six countries met the inclusion criteria. These criteria required histologically confirmed GEA in the metastatic or unresectable setting with EGFR amplification identified by using a Clinical Laboratory Improvement Amendments–approved assay, and who received on- or off-protocol EGFRi. Testing could be by tissue next-generation sequencing, plasma circulating tumor DNA next-generation sequencing, and/or fluorescence in situ hybridization performed by a Clinical Laboratory Improvement Amendments approved laboratory. Treatment patterns and outcomes analysis was also performed using a deidentified clinicogenomic database (CGDB). RESULTS Sixty patients with EGFR-amplified GEA received EGFRi, including 31 of 60 patients (52%) with concurrent chemotherapy. Across treatment lines, patients achieved a 43% objective response rate with a median progression-free survival of 4.6 months (95% CI, 3.5 to 6.4). Patients receiving EGFRi in first-, second-, and third-line therapy achieved a median overall survival of 20.6 months (95% CI, 13.5 to not reached [NR]), 9 months (95% CI, 7.9 to NR), and 8.4 months (7.6 to NR), respectively. This survival far exceeded the 11.2-month (95% CI, 8.7 to 14.2) median overall survival from first-line initiation of non-EGFRi therapy in patients with EGFR-amplified GEA in the CGDB. Despite this benefit, analysis of the CGDB (January 2011-December 2020) suggests that only 5% of patients with EGFR-amplified GEA received EGFRi. CONCLUSION Patients with EGFR-amplified GEA derive significant benefit from EGFRi. Further prospective investigation of EGFRi in a well-selected patient population is ongoing in an upcoming trial of amivantamab in EGFR and/or MET amplified GEA.
- Published
- 2023
5. PD-L1 expression and overall survival in Asian and western patients with gastric cancer
- Author
-
Sun Young Rha, Geoffrey Y Ku, Hyo Song Kim, Hyun Cheol Chung, Fatemeh Ghazanfari Amlashi, Dipen Maheshbhai Maru, Carly A Fein, Laura H Tang, Wei Zhou, Ting Wu, Senaka A Peter, David P Kelsen, and Jaffer A Ajani
- Subjects
Cancer Research ,Oncology ,Stomach Neoplasms ,Biomarkers, Tumor ,Humans ,General Medicine ,B7-H1 Antigen ,Retrospective Studies - Abstract
Aim: Data are limited on PD-L1 expression and its association with overall survival (OS) in gastric cancer (GC) patients receiving routine care in different regions. Materials & methods: In a retrospective study, PD-L1 expression was assayed using the 22C3 pharmDx on GC tumor samples collected between 2003 and 2017 at South Korean and US cancer centers. PD-L1 positivity was defined as combined positive score (CPS) ≥1. The relationship between PD-L1 and OS was analyzed. Results: Of 574 GC tumor samples, 67.4% were CPS ≥1 (68.7% in Korean and 65.7% in US patients). PD-L1 expression was not associated with OS (adjusted hazard ratio: 0.94; 95% CI: 0.75–1.17). Conclusion: PD-L1 prevalence and its association with OS was similar between South Korean and US GC patients.
- Published
- 2022
- Full Text
- View/download PDF
6. Survival of Locally Advanced MSI-high Gastric Cancer Patients Treated With Perioperative Chemotherapy
- Author
-
Elvira L, Vos, Steven B, Maron, Robert W, Krell, Masaya, Nakauchi, Megan, Fiasconaro, Marinela, Capanu, Henry S, Walch, Walid K, Chatila, Nikolaus, Schultz, David H, Ilson, Yelena Y, Janjigian, Geoffrey Y, Ku, Sam S, Yoon, Daniel G, Coit, Chad M, Vanderbilt, Laura H, Tang, and Vivian E, Strong
- Subjects
Surgery - Abstract
To evaluate the efficacy of chemotherapy in patients with microsatellite instability-high (MSI-high) gastric cancer.Although MSI-high gastric cancer is associated with superior prognosis, recent studies question the benefit of perioperative chemotherapy in this population.Locally advanced gastric adenocarcinoma patients who either underwent surgery alone or also received neoadjuvant, perioperative, or adjuvant chemotherapy between 2000-2018 were eligible. MSI status, determined by next generation sequencing or mismatch repair protein immunohistochemistry, was determined in 535 patients. Associations among MSI status, chemotherapy administration, and overall survival (OS), disease-specific survival (DSS), and disease-free survival (DFS) were assessed.In 535 patients, 82 (15.3%) had an MSI-high tumor and approximately 20% better OS, DSS, and DFS. Grade 1 (90-100%) pathological response to neoadjuvant chemotherapy was found in 0 of 40 (0%) MSI-high tumors versus 43 of 274 (16%) MSS. In the MSI-high group, the 3-year OS rate was 79% with chemotherapy versus 88% with surgery alone (P=0.48). In the MSS group, this was 61% versus 59%, respectively (P=0.96). After multivariable interaction analyses, patients with MSI-high tumors had superior survival compared with patients with MSS tumors whether given chemotherapy (HR 0.53, 95% CI 0.28-0.99) or treated with surgery alone (HR 0.15 vs. MSS, 95% CI 0.02-1.17).MSI-high locally advanced gastric cancer was associated with superior survival compared to MSS overall, despite worse pathological chemotherapy response. In patients with MSI-high gastric cancer who received chemotherapy, the survival rate was approximately 9% worse compared with surgery alone, but chemotherapy was not significantly associated with survival.
- Published
- 2022
- Full Text
- View/download PDF
7. Approach to Resectable Gastric Cancer: Evolving Paradigm of Neoadjuvant and Adjuvant Treatment
- Author
-
Melissa A. Lumish and Geoffrey Y. Ku
- Subjects
Oncology ,Pharmacology (medical) - Published
- 2022
- Full Text
- View/download PDF
8. The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Author
-
Smita Sihag, Samuel C. Nussenzweig, Henry S. Walch, Meier Hsu, Kay See Tan, Sergio De La Torre, Yelena Y. Janjigian, Steven B. Maron, Geoffrey Y. Ku, Laura H. Tang, Pari M. Shah, Abraham Wu, David R. Jones, David B. Solit, Nikolaus Schultz, Karuna Ganesh, Michael F. Berger, and Daniela Molena
- Subjects
Cancer Research ,Esophageal Neoplasms ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Prospective Studies ,Adenocarcinoma ,Tumor Suppressor Protein p53 ,Neoadjuvant Therapy ,Article ,Retrospective Studies - Abstract
Purpose: In patients with locally advanced esophageal adenocarcinoma, response to neoadjuvant therapy strongly predicts survival, but robust molecular predictors of response have been lacking. We therefore sought to discover meaningful predictors of response in these patients. Experimental Design: We retrospectively identified all patients with adenocarcinoma of the lower esophagus or gastroesophageal junction who (i) were treated with multimodality therapy with curative intent at our institution from 2014 through 2020 and (ii) underwent prospective sequencing by Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets. Clinicopathologic and genomic data were analyzed to identify potential genomic features, somatic alterations, and oncogenic pathways associated with treatment response. Results: In total, 237 patients were included. MDM2 amplification was independently associated with poor response to neoadjuvant therapy [OR, 0.10 (95% confidence interval, 0.01–0.55); P = 0.032], when accounting for significant clinicopathologic variables, including clinical stage, tumor grade, and chemotherapy regimen. Moreover, TP53 pathway alterations, grouped according to inferred severity of TP53 dysfunction, were significantly associated with response to neoadjuvant therapy (P = 0.004, q = 0.07). Patients with MDM2 amplifications or truncating biallelic TP53 mutations had similar outcomes in terms of poor responses to neoadjuvant therapy and, consequently, shorter progression-free survival, compared with patients with TP53 pathway wild-type tumors. Thus, worsening TP53 dysfunction was directly correlated with worse outcomes. Conclusions: MDM2 amplification and TP53 status are associated with response to therapy in patients with esophageal adenocarcinoma. Given the dearth of actionable targets in esophageal adenocarcinoma, MDM2 inhibition, in combination with cytotoxic chemotherapy, may represent an important therapeutic strategy to overcome treatment resistance and improve outcomes in these patients.
- Published
- 2022
- Full Text
- View/download PDF
9. Data from Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer
- Author
-
Nikolaus Schultz, David B. Solit, Barry S. Taylor, Laura Tang, Michael F. Berger, David H. Ilson, Neal Rosen, Marc Ladanyi, Maurizio Scaltriti, David M. Hyman, Ahmet Zehir, Sumit Middha, Marinela Capanu, Mark E. Robson, Jinru Shia, Daniela Molena, David R. Jones, Valerie W. Rusch, Zsofia K. Stadler, Manjit Bains, Daniel G. Coit, Hans Gerdes, Mark Schattner, Vivian E. Strong, Liying Zhang, David P. Kelsen, Benjamin E. Gross, Zachary J. Heins, Jianjiong Gao, Efsevia Vakiani, Nancy Bouvier, Ritika Kundra, Yaelle Tuvy, Jamie C. Riches, Geoffrey Y. Ku, Jaclyn F. Hechtman, Walid K. Chatila, Philip Jonsson, Francisco Sanchez-Vega, and Yelena Y. Janjigian
- Abstract
The incidence of esophagogastric cancer is rapidly rising, but only a minority of patients derive durable benefit from current therapies. Chemotherapy as well as anti-HER2 and PD-1 antibodies are standard treatments. To identify predictive biomarkers of drug sensitivity and mechanisms of resistance, we implemented prospective tumor sequencing of patients with metastatic esophagogastric cancer. There was no association between homologous recombination deficiency defects and response to platinum-based chemotherapy. Patients with microsatellite instability–high tumors were intrinsically resistant to chemotherapy but more likely to achieve durable responses to immunotherapy. The single Epstein–Barr virus–positive patient achieved a durable, complete response to immunotherapy. The level of ERBB2 amplification as determined by sequencing was predictive of trastuzumab benefit. Selection for a tumor subclone lacking ERBB2 amplification, deletion of ERBB2 exon 16, and comutations in the receptor tyrosine kinase, RAS, and PI3K pathways were associated with intrinsic and/or acquired trastuzumab resistance. Prospective genomic profiling can identify patients most likely to derive durable benefit to immunotherapy and trastuzumab and guide strategies to overcome drug resistance.Significance: Clinical application of multiplex sequencing can identify biomarkers of treatment response to contemporary systemic therapies in metastatic esophagogastric cancer. This large prospective analysis sheds light on the biological complexity and the dynamic nature of therapeutic resistance in metastatic esophagogastric cancers. Cancer Discov; 8(1); 49–58. ©2017 AACR.See related commentary by Sundar and Tan, p. 14.See related article by Pectasides et al., p. 37.This article is highlighted in the In This Issue feature, p. 1
- Published
- 2023
- Full Text
- View/download PDF
10. Figure S2 from Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer
- Author
-
Nikolaus Schultz, David B. Solit, Barry S. Taylor, Laura Tang, Michael F. Berger, David H. Ilson, Neal Rosen, Marc Ladanyi, Maurizio Scaltriti, David M. Hyman, Ahmet Zehir, Sumit Middha, Marinela Capanu, Mark E. Robson, Jinru Shia, Daniela Molena, David R. Jones, Valerie W. Rusch, Zsofia K. Stadler, Manjit Bains, Daniel G. Coit, Hans Gerdes, Mark Schattner, Vivian E. Strong, Liying Zhang, David P. Kelsen, Benjamin E. Gross, Zachary J. Heins, Jianjiong Gao, Efsevia Vakiani, Nancy Bouvier, Ritika Kundra, Yaelle Tuvy, Jamie C. Riches, Geoffrey Y. Ku, Jaclyn F. Hechtman, Walid K. Chatila, Philip Jonsson, Francisco Sanchez-Vega, and Yelena Y. Janjigian
- Abstract
Concordance between copy-number inferred from next generation sequencing and protein overexpression by IHC or gene amplification by FISH.
- Published
- 2023
- Full Text
- View/download PDF
11. Figure S1 from EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer
- Author
-
Yelena Y. Janjigian, Nikolaus Schultz, Maurizio Scaltriti, Jorge A. Carrasquillo, David B. Solit, Jason S. Lewis, Barry S. Taylor, Elisa de Stanchina, Michael F. Berger, Wolfgang A. Weber, David H. Ilson, Christine A. Iacobuzio-Donahue, Heiko Schöder, Neeta Pandit-Taskar, Steven M. Larson, Richard B. Lanman, Rebecca J. Nagy, Todd Hembrough, Yuan Tian, Fabiola Cecchi, Besnik Qeriqi, Marissa Mattar, Marinela Capanu, Mario E. Lacouture, Karen Brown, Robert A. Lefkowitz, David P. Kelsen, Mark Schattner, Efsevia Vakiani, Joanne Soong, Gouri J. Nanjangud, Nancy Bouvier, Christopher J. Fong, Helen Won, Yaelle Tuvy, Geoffrey Y. Ku, Pau Castel, Jaclyn F. Hechtman, and Francisco Sanchez-Vega
- Abstract
Additional results for dual probe EGFR and ERBB2 FISH from Patients 9, 30 and 32.
- Published
- 2023
- Full Text
- View/download PDF
12. Supplementary Figure Legends from Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer
- Author
-
Nikolaus Schultz, David B. Solit, Barry S. Taylor, Laura Tang, Michael F. Berger, David H. Ilson, Neal Rosen, Marc Ladanyi, Maurizio Scaltriti, David M. Hyman, Ahmet Zehir, Sumit Middha, Marinela Capanu, Mark E. Robson, Jinru Shia, Daniela Molena, David R. Jones, Valerie W. Rusch, Zsofia K. Stadler, Manjit Bains, Daniel G. Coit, Hans Gerdes, Mark Schattner, Vivian E. Strong, Liying Zhang, David P. Kelsen, Benjamin E. Gross, Zachary J. Heins, Jianjiong Gao, Efsevia Vakiani, Nancy Bouvier, Ritika Kundra, Yaelle Tuvy, Jamie C. Riches, Geoffrey Y. Ku, Jaclyn F. Hechtman, Walid K. Chatila, Philip Jonsson, Francisco Sanchez-Vega, and Yelena Y. Janjigian
- Abstract
Text legends for Supplementary Figures S1-S4.
- Published
- 2023
- Full Text
- View/download PDF
13. Tables S1-S5 from Genetic Predictors of Response to Systemic Therapy in Esophagogastric Cancer
- Author
-
Nikolaus Schultz, David B. Solit, Barry S. Taylor, Laura Tang, Michael F. Berger, David H. Ilson, Neal Rosen, Marc Ladanyi, Maurizio Scaltriti, David M. Hyman, Ahmet Zehir, Sumit Middha, Marinela Capanu, Mark E. Robson, Jinru Shia, Daniela Molena, David R. Jones, Valerie W. Rusch, Zsofia K. Stadler, Manjit Bains, Daniel G. Coit, Hans Gerdes, Mark Schattner, Vivian E. Strong, Liying Zhang, David P. Kelsen, Benjamin E. Gross, Zachary J. Heins, Jianjiong Gao, Efsevia Vakiani, Nancy Bouvier, Ritika Kundra, Yaelle Tuvy, Jamie C. Riches, Geoffrey Y. Ku, Jaclyn F. Hechtman, Walid K. Chatila, Philip Jonsson, Francisco Sanchez-Vega, and Yelena Y. Janjigian
- Abstract
Supplementary Table 1: Comprehensive clinical data and sample IDs. Supplementary Table 2: Summary of baseline patient and sample characteristics. Supplementary Table 3: List of somatic mutations identified in all samples. Supplementary Table 4: Detailed data for assesment of ERBB2 amplification. Supplementary Table 5: Genes included on MSK-IMPACT platform
- Published
- 2023
- Full Text
- View/download PDF
14. Supplementary Figure Legends from EGFR and MET Amplifications Determine Response to HER2 Inhibition in ERBB2-Amplified Esophagogastric Cancer
- Author
-
Yelena Y. Janjigian, Nikolaus Schultz, Maurizio Scaltriti, Jorge A. Carrasquillo, David B. Solit, Jason S. Lewis, Barry S. Taylor, Elisa de Stanchina, Michael F. Berger, Wolfgang A. Weber, David H. Ilson, Christine A. Iacobuzio-Donahue, Heiko Schöder, Neeta Pandit-Taskar, Steven M. Larson, Richard B. Lanman, Rebecca J. Nagy, Todd Hembrough, Yuan Tian, Fabiola Cecchi, Besnik Qeriqi, Marissa Mattar, Marinela Capanu, Mario E. Lacouture, Karen Brown, Robert A. Lefkowitz, David P. Kelsen, Mark Schattner, Efsevia Vakiani, Joanne Soong, Gouri J. Nanjangud, Nancy Bouvier, Christopher J. Fong, Helen Won, Yaelle Tuvy, Geoffrey Y. Ku, Pau Castel, Jaclyn F. Hechtman, and Francisco Sanchez-Vega
- Abstract
Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
15. Supplementary Data from The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Author
-
Daniela Molena, Michael F. Berger, Karuna Ganesh, Nikolaus Schultz, David B. Solit, David R. Jones, Abraham Wu, Pari M. Shah, Laura H. Tang, Geoffrey Y. Ku, Steven B. Maron, Yelena Y. Janjigian, Sergio De La Torre, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Supplementary Data from The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Published
- 2023
- Full Text
- View/download PDF
16. Data from The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Author
-
Daniela Molena, Michael F. Berger, Karuna Ganesh, Nikolaus Schultz, David B. Solit, David R. Jones, Abraham Wu, Pari M. Shah, Laura H. Tang, Geoffrey Y. Ku, Steven B. Maron, Yelena Y. Janjigian, Sergio De La Torre, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Purpose:In patients with locally advanced esophageal adenocarcinoma, response to neoadjuvant therapy strongly predicts survival, but robust molecular predictors of response have been lacking. We therefore sought to discover meaningful predictors of response in these patients.Experimental Design:We retrospectively identified all patients with adenocarcinoma of the lower esophagus or gastroesophageal junction who (i) were treated with multimodality therapy with curative intent at our institution from 2014 through 2020 and (ii) underwent prospective sequencing by Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets. Clinicopathologic and genomic data were analyzed to identify potential genomic features, somatic alterations, and oncogenic pathways associated with treatment response.Results:In total, 237 patients were included. MDM2 amplification was independently associated with poor response to neoadjuvant therapy [OR, 0.10 (95% confidence interval, 0.01–0.55); P = 0.032], when accounting for significant clinicopathologic variables, including clinical stage, tumor grade, and chemotherapy regimen. Moreover, TP53 pathway alterations, grouped according to inferred severity of TP53 dysfunction, were significantly associated with response to neoadjuvant therapy (P = 0.004, q = 0.07). Patients with MDM2 amplifications or truncating biallelic TP53 mutations had similar outcomes in terms of poor responses to neoadjuvant therapy and, consequently, shorter progression-free survival, compared with patients with TP53 pathway wild-type tumors. Thus, worsening TP53 dysfunction was directly correlated with worse outcomes.Conclusions:MDM2 amplification and TP53 status are associated with response to therapy in patients with esophageal adenocarcinoma. Given the dearth of actionable targets in esophageal adenocarcinoma, MDM2 inhibition, in combination with cytotoxic chemotherapy, may represent an important therapeutic strategy to overcome treatment resistance and improve outcomes in these patients.
- Published
- 2023
- Full Text
- View/download PDF
17. Data from Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways
- Author
-
Michael F. Berger, Nikolaus Schultz, David B. Solit, Daniela Molena, David R. Jones, Abraham J. Wu, Pari M. Shah, Jaclyn Hechtman, Laura H. Tang, Geoffrey Y. Ku, Steven Maron, Yelena Y. Janjigian, Assem Patel, Sergio A. De La Torre, Walid K. Chatila, Francisco Sanchez-Vega, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Purpose:To delineate recurrent oncogenic driver alterations and dysregulated pathways in esophageal adenocarcinoma and to assess their prognostic value.Experimental Design:We analyzed a large cohort of patients with lower esophageal and junctional adenocarcinoma, prospectively sequenced by MSK-IMPACT with high-quality clinical annotation. Patients were subdivided according to treatment intent, curative versus palliative, which closely mirrored clinical staging. Genomic features, alterations, and pathways were examined for association with overall survival using Cox proportional hazard models, adjusted for relevant clinicopathologic factors knowable at the time of diagnosis.Results:Analysis of 487 patients revealed 16 oncogenic driver alterations, mostly amplifications, present in ≥5% of patients. Patients in the palliative-intent cohort, compared with those in the curative-intent cohort, were more likely to have metastatic disease, ERBB2 amplifications, Cell-cycle and RTK–RAS pathway alterations, as well as a higher fraction of genome altered and rate of whole-genome doubling. In multivariable analyses, CDKN2A alterations, SMAD4 alterations, KRAS amplifications, Cell-cycle and TGFβ pathways, and overall number of oncogenic drivers were independently associated with worse overall survival. ERBB2 amplification was associated with improved survival, presumably due to trastuzumab therapy.Conclusions:Our study suggests that higher levels of genomic instability are associated with more advanced disease in esophageal adenocarcinoma. Furthermore, CDKN2A, KRAS, and SMAD4 represent prognostic biomarkers, given their strong association with poor survival.
- Published
- 2023
- Full Text
- View/download PDF
18. Supplementary Figure 1 from Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways
- Author
-
Michael F. Berger, Nikolaus Schultz, David B. Solit, Daniela Molena, David R. Jones, Abraham J. Wu, Pari M. Shah, Jaclyn Hechtman, Laura H. Tang, Geoffrey Y. Ku, Steven Maron, Yelena Y. Janjigian, Assem Patel, Sergio A. De La Torre, Walid K. Chatila, Francisco Sanchez-Vega, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Alterations in esophageal adenocarcinoma in the TCGA dataset, and mapping of alterations onto the RTK-RAS and Cell Cycle pathways.
- Published
- 2023
- Full Text
- View/download PDF
19. Supplementary Figure from The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Author
-
Daniela Molena, Michael F. Berger, Karuna Ganesh, Nikolaus Schultz, David B. Solit, David R. Jones, Abraham Wu, Pari M. Shah, Laura H. Tang, Geoffrey Y. Ku, Steven B. Maron, Yelena Y. Janjigian, Sergio De La Torre, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Supplementary Figure from The Role of the TP53 Pathway in Predicting Response to Neoadjuvant Therapy in Esophageal Adenocarcinoma
- Published
- 2023
- Full Text
- View/download PDF
20. Supplementary Tables from Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways
- Author
-
Michael F. Berger, Nikolaus Schultz, David B. Solit, Daniela Molena, David R. Jones, Abraham J. Wu, Pari M. Shah, Jaclyn Hechtman, Laura H. Tang, Geoffrey Y. Ku, Steven Maron, Yelena Y. Janjigian, Assem Patel, Sergio A. De La Torre, Walid K. Chatila, Francisco Sanchez-Vega, Kay See Tan, Meier Hsu, Henry S. Walch, Samuel C. Nussenzweig, and Smita Sihag
- Abstract
Supplementary Tables 1 and 2
- Published
- 2023
- Full Text
- View/download PDF
21. Supplementary Data from Efficacy of Combined VEGFR1-3, PDGFα/β, and FGFR1-3 Blockade Using Nintedanib for Esophagogastric Cancer
- Author
-
Yelena Y. Janjigian, Nikolaus Schultz, David B. Solit, Marinela Capanu, David H. Ilson, Mark Schattner, David P. Kelsen, Laura Tang, Michael F. Berger, Yaelle Tuvy, Avni M. Desai, Zoe Goldberg, Michelle S. Boyar, Sree B. Chalasani, Geoffrey Y. Ku, Joanne F. Chou, Jaclyn F. Hechtman, Walid K. Chatila, Azfar Basunia, and Elizabeth Won
- Abstract
Supplementary Table 1
- Published
- 2023
- Full Text
- View/download PDF
22. Data from Clinical and Molecular Predictors of Response to Immune Checkpoint Inhibitors in Patients with Advanced Esophagogastric Cancer
- Author
-
Geoffrey Y. Ku, Yelena Y. Janjigian, Nikolaus Schultz, Luis A. Diaz, David B. Solit, Laura H. Tang, Michael F. Berger, David H. Ilson, David P. Kelsen, Marc Ladanyi, Foysal Daian, Ritika Kundra, Yaelle Tuvy, Jaclyn F. Hechtman, Marinela Capanu, Matthew Margolis, Walid K. Chatila, Joanne F. Chou, and Megan Greally
- Abstract
Purpose:Immune checkpoint inhibitors (ICI) are effective in only a minority of patients with esophagogastric cancer (EGC). Here, we aimed to identify predictors of durable clinical benefit to ICI in EGC.Experimental Design:Patients with advanced EGC treated with ICIs at Memorial Sloan Kettering Cancer Center (New York, NY) were identified. Clinicopathologic variables were assessed. In patients profiled by MSK-IMPACT–targeted sequencing, outcomes were correlated with tumor genomic features.Results:One-hundred sixty-one patients were treated with ICIs (110 with anti–PD-1/PD-L1 antibodies and 51 with anti–CTLA-4 and PD-1/PD-L1 antibodies). The median progression-free survival (PFS) and overall survival (OS) were 1.7 and 4.9 months, respectively. Greater number of disease sites (≥3), liver metastases, treatment with ≥3 prior therapies and ECOG performance status ≥2 were associated with poorer PFS and OS. Patients treated with combination ICI and those with PD-L1–positive tumors had improved outcomes. There was no difference in outcomes between patients treated with antibiotics during or in the 2 months preceding ICI treatment versus those who were not. Occurrence of irAEs was associated with improved OS. In genomically profiled tumors (n = 89), survival was associated with increasing tumor mutation burden (TMB). However, in multivariable analyses and when microsatellite unstable (MSI) patients were excluded, a significant association was no longer observed.Conclusions:In patients with advanced EGC, heavily pretreated patients, those with high-volume disease and/or poor PS were less likely to benefit from ICI. irAEs were associated with improved OS. TMB correlated with improved survival, but this association was not observed when MSI-high patients were excluded.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary Data from Clinical and Molecular Predictors of Response to Immune Checkpoint Inhibitors in Patients with Advanced Esophagogastric Cancer
- Author
-
Geoffrey Y. Ku, Yelena Y. Janjigian, Nikolaus Schultz, Luis A. Diaz, David B. Solit, Laura H. Tang, Michael F. Berger, David H. Ilson, David P. Kelsen, Marc Ladanyi, Foysal Daian, Ritika Kundra, Yaelle Tuvy, Jaclyn F. Hechtman, Marinela Capanu, Matthew Margolis, Walid K. Chatila, Joanne F. Chou, and Megan Greally
- Abstract
PFS analysis by gene alteration occurring in 10% or more of MSS patients who received combination therapy
- Published
- 2023
- Full Text
- View/download PDF
24. ATM Germline-Mutated Gastroesophageal Junction Adenocarcinomas: Clinical Descriptors, Molecular Characteristics, and Potential Therapeutic Implications
- Author
-
Tony El Jabbour, Maksym Misyura, Darren Cowzer, Michal Zimmermann, Victoria Rimkunas, Antonio Marra, Fatemeh Derakhshan, Pier Selenica, Megan Parilla, Jeremy S Setton, Ozge Ceyhan-Birsoy, Yelena Kemel, Amanda Catchings, Megha Ranganathan, Geoffrey Y Ku, Yelena Y Janjigian, Michael Zinda, Maria Koehler, Zsofia Stadler, Jinru Shia, Jorge S Reis-Filho, and Diana Mandelker
- Subjects
Cancer Research ,Germ Cells ,Esophageal Neoplasms ,Oncology ,Stomach Neoplasms ,Humans ,Ataxia Telangiectasia Mutated Proteins ,Esophagogastric Junction ,Adenocarcinoma - Abstract
Background Gastroesophageal junction (GEJ) adenocarcinoma is a rare cancer associated with poor prognosis. The genetic factors conferring predisposition to GEJ adenocarcinoma have yet to be identified. Methods We analyzed germline testing results from 23 381 cancer patients undergoing tumor-normal sequencing, of which 312 individuals had GEJ adenocarcinoma. Genomic profiles and clinico-pathologic features were analyzed for the GEJ adenocarcinomas. Silencing of ATM and ATR was performed using validated short-interfering RNA species in GEJ, esophageal, and gastric adenocarcinoma cell lines. All statistical tests were 2-sided. Results Pathogenic or likely pathogenic ATM variants were identified in 18 of 312 patients (5.8%), and bi-allelic inactivation of ATM through loss of heterozygosity of the wild-type allele was detected in all (16 of 16) samples with sufficient tumor content. Germline ATM-mutated GEJ adenocarcinomas largely lacked somatic mutations in TP53, were more likely to harbor MDM2 amplification, and harbored statistically significantly fewer somatic single nucleotide variants (2.0 mutations/Mb vs 7.9 mutations/Mb; P < .001). A statistically significantly higher proportion of germline ATM-mutated than ATM–wild-type GEJ adenocarcinoma patients underwent a curative resection (10 [100%] vs 92 [86.8%], P = .04; Fisher’s exact test.), A synthetic lethal interaction between short-interfering RNA silencing of ATM and ATR was observed in the models analyzed. Conclusions Our results indicate that germline pathogenic variants in ATM drive oncogenesis in GEJ adenocarcinoma and might result in a distinct clinical phenotype. Given the high prevalence of germline ATM-mutated GEJ adenocarcinomas, genetic testing for individuals with GEJ adenocarcinomas may be considered to better inform prognostication, treatment decisions, and future cancer risk.
- Published
- 2022
- Full Text
- View/download PDF
25. Phase I/Ib study of crenolanib with ramucirumab and paclitaxel as second-line therapy for advanced esophagogastric adenocarcinoma
- Author
-
Ryan H. Moy, Megan Greally, Joanne F. Chou, Jia Li, Avni M. Desai, Sree B. Chalasani, Elizabeth Won, David P. Kelsen, David H. Ilson, Yelena Y. Janjigian, Marinela Capanu, and Geoffrey Y. Ku
- Subjects
Pharmacology ,Cancer Research ,Oncology ,Pharmacology (medical) ,Toxicology - Published
- 2022
- Full Text
- View/download PDF
26. A nutritional management algorithm in older patients with locally advanced esophageal cancer
- Author
-
David R. Jones, Daniela Molena, Geoffrey Y. Ku, William P. Tew, Steven Brad Maron, Arlyn Apollo, Yelena Y. Janjigian, Shalom Sabwa, David H. Ilson, Ryan H. Moy, Elizabeth Won, Abraham J. Wu, and Marina Shcherba
- Subjects
medicine.medical_specialty ,Chemotherapy ,Esophageal Neoplasms ,business.industry ,medicine.medical_treatment ,Locally advanced ,Nutritional status ,Chemoradiotherapy ,Esophageal cancer ,medicine.disease ,Article ,Management algorithm ,Oncology ,Older patients ,Geriatric oncology ,Quality of life ,medicine ,Humans ,sense organs ,Geriatrics and Gerontology ,skin and connective tissue diseases ,Intensive care medicine ,business ,Algorithms ,Aged - Abstract
e16038Background: Esophageal cancer primarily affects older adults, who are at highest risk for poor nutritional status due to medical comorbidities, physiological changes of aging and geriatric is...
- Published
- 2022
- Full Text
- View/download PDF
27. Oligometastases After Curative Esophagectomy Are Not One Size Fits All
- Author
-
David R. Jones, Kay See Tan, Mahmoud Eljalby, Daniela Molena, Abraham J. Wu, Yelena Y. Janjigian, Smita Sihag, Geoffrey Y. Ku, Tamar B. Nobel, Meier Hsu, David B. Sewell, Manjit S. Bains, and Xin Xin Xing
- Subjects
Male ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Systemic disease ,Lung Neoplasms ,Esophageal Neoplasms ,medicine.medical_treatment ,Adenocarcinoma ,Gastroenterology ,Disease-Free Survival ,Article ,Internal medicine ,Overall survival ,Humans ,Medicine ,Neoplasm Metastasis ,Aged ,Neoplasm Staging ,Retrospective Studies ,Lung ,Brain Neoplasms ,business.industry ,Proportional hazards model ,Liver Neoplasms ,Middle Aged ,Esophageal cancer ,Prognosis ,medicine.disease ,United States ,Confidence interval ,Esophagectomy ,Survival Rate ,medicine.anatomical_structure ,Female ,Surgery ,Neoplasm Recurrence, Local ,Cardiology and Cardiovascular Medicine ,business - Abstract
BACKGROUND: More than half of patients treated with esophagectomy for esophageal cancer experience recurrence. Oligometastasis, a proposed intermediate state of isolated local or solid-organ recurrence that occurs before widespread systemic disease, is a potential target for aggressive local intervention. We investigated presentation and prognosis between solid-organ recurrence sites. METHODS: Patients with isolated solid-organ recurrence at the liver, lung, or brain who underwent R0 esophagectomy from 1995 to 2016 were identified. Clinicopathologic characteristics and outcomes were compared between sites of recurrence. Overall survival was quantified using the Kaplan-Meier approach and Cox proportional hazards models. RESULTS: In total, 104 patients were included (site: brain, 37; lung, 27; liver, 40). Eighty percent of liver, 51% of brain, and 44% of lung oligometastases occurred in the first 12 months after esophagectomy. Despite the limited use of aggressive therapy, patients with lung oligometastasis had significantly longer median overall survival (2.41 years; 95% confidence interval [CI], 1.58–3.31) than patients with brain (0.95 years; 95% CI, 0.62–1.49) or liver (0.95 years; 95% CI, 0.82–1.41) oligometastasis (p
- Published
- 2021
- Full Text
- View/download PDF
28. Durvalumab and pet-directed chemoradiation in locally advanced esophageal adenocarcinoma – a phase ib/ii study
- Author
-
Darren Cowzer, Abraham Jing-Ching Wu, Smita Sihag, Henry S. Walch, Bernard J. Park, David Randolph Jones, Ping Gu, Steven B. Maron, Ryan Sugarman, Sree Bhavani Chalasani, Marina Shcherba, Marinela Capanu, Joanne F. Chou, Jennie K Choe, Anton Nosov, Prasad S Adusumilli, Randy Yeh, Laura H. Tang, David H. Ilson, Yelena Y. Janjigian, Daniela Molena, and Geoffrey Y. Ku
- Subjects
Surgery - Published
- 2023
- Full Text
- View/download PDF
29. Incidence and Management of Esophageal Cancer Recurrence to Regional Lymph Nodes After Curative Esophagectomy
- Author
-
Thomas Boerner, Rebecca A. Carr, Meier Hsu, Alexa Michel, Kay See Tan, Elvira Vos, Smita Sihag, Manjit S. Bains, Geoffrey Y. Ku, Abraham J. Wu, David R. Jones, and Daniela Molena
- Subjects
Cancer Research ,Oncology - Abstract
Up to 50% of patients treated with curative esophagectomy for esophageal cancer will develop recurrence, contributing to the dismal survival associated with this disease. Regional recurrence may represent disease that is not yet widely metastatic and may therefore be amenable to more-aggressive treatment. We sought to assess all patients treated with curative esophagectomy for esophageal cancer who developed regional recurrence. We retrospectively identified all patients who underwent esophagectomy for esophageal adenocarcinoma and esophageal squamous cell carcinoma at a single institution from January 2000 to August 2019. In total, 1626 patients were included in the study cohort. As of June 2022, 595 patients had disease recurrence, which was distant or systemic in 435 patients (27%), regional in 125 (7.7%), and local in 35 (2.2%). On multivariable analysis, neoadjuvant chemoradiation with a total radiation dose45 Gy (hazard ratio [HR], 3.5 [95% CI, 1.7-7.3]; P=0.001), pathologic node-positive disease (HR, 1.9 [95% CI, 1.3-3.0]; P=0.003), and lymphovascular invasion (HR, 1.6 [95% CI, 1.0-2.5]; P=0.049) were predictors of isolated nodal recurrence, whereas increasing age (HR, 0.97 [95% CI, 0.96-0.99]; P=0.001) and increasing number of excised lymph nodes (HR, 0.98 [95% CI, 0.95-1.00]; P=0.021) were independently associated with decreased risk of regional recurrence. Patients treated with a combination of local and systemic therapies had better survival outcomes than patients treated with systemic therapy alone (P0.001). In patients with recurrence of esophageal cancer limited to regional lymph nodes, salvage treatment may be possible. Higher radiation doses and more-extensive lymphadenectomy may reduce the risk of regional recurrence. This article is protected by copyright. All rights reserved.
- Published
- 2022
30. Induction FOLFOX and PET-Directed Chemoradiation for Locally Advanced Esophageal Adenocarcinoma
- Author
-
David H. Ilson, Smita Sihag, Manjit S. Bains, Geoffrey Y. Ku, Valerie W. Rusch, Meier Hsu, Abraham J. Wu, Rebecca Carr, Matthew J. Bott, Caitlin Harrington, Steven B Maron, David R. Jones, Daniela Molena, James M. Isbell, Yelena Y. Janjigian, Bernard J. Park, and Kay See Tan
- Subjects
medicine.medical_specialty ,Proportional hazards model ,business.industry ,medicine.medical_treatment ,Standard treatment ,Induction chemotherapy ,Cancer ,medicine.disease ,Gastroenterology ,Carboplatin ,Exact test ,chemistry.chemical_compound ,FOLFOX ,chemistry ,Esophagectomy ,Internal medicine ,medicine ,Surgery ,business ,medicine.drug - Abstract
OBJECTIVE To compare the efficacy and safety of induction FOLFOX followed by PET-directed neoadjuvant chemoradiation therapy (nCRT), induction carboplatin plus paclitaxel (CP) followed by PET-directed nCRT, and nCRT with CP alone in patients with esophageal adenocarcinoma (EAC). SUMMARY BACKGROUND DATA nCRT with CP is a standard treatment for locally advanced EAC. The results of Cancer and Leukemia Group B 80803 support the use of induction chemotherapy followed by PET-directed chemoradiation therapy. METHODS We retrospectively identified all patients with EAC who underwent the treatments above followed by esophagectomy. We assessed incidences of pathologic complete response (pCR), near-pCR (ypN0 with ≥90% response), and surgical complications between treatment groups using Fisher's exact test and logistic regression; disease-free survival (DFS) and overall survival (OS) were estimated by the Kaplan-Meier method and evaluated using the log-rank test and extended Cox regression. RESULTS In total, 451 patients were included: 309 (69%) received induction chemotherapy before nCRT (FOLFOX, n=70; CP, n=239); 142 (31%) received nCRT with CP. Rates of pCR (33% vs 16%, P=0.004), near-pCR (57% vs 33%, P
- Published
- 2021
- Full Text
- View/download PDF
31. Comparison of Long- and Short-term Outcomes in 845 Open and Minimally Invasive Gastrectomies for Gastric Cancer in the United States
- Author
-
Daniel G. Coit, Makoto Nishimura, Vivian E. Strong, Geoffrey Y. Ku, Masaya Nakauchi, Mithat Gonen, Yelena Y. Janjigian, Elvira L. Vos, and Mark A. Schattner
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Article ,03 medical and health sciences ,0302 clinical medicine ,Robotic Surgical Procedures ,Gastrectomy ,Stomach Neoplasms ,Surgical oncology ,medicine ,Humans ,Minimally Invasive Surgical Procedures ,Retrospective Studies ,Surgical approach ,business.industry ,Open surgery ,Hazard ratio ,Cancer ,Odds ratio ,Length of Stay ,medicine.disease ,United States ,Surgery ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Cohort ,Laparoscopy ,030211 gastroenterology & hepatology ,business - Abstract
Few Western studies have evaluated the long-term oncologic outcomes of minimally invasive surgery (MIS) approaches to gastrectomy for gastric cancer. This study aimed to compare the outcomes between minimally invasive and open gastrectomies and between laparoscopic and robotic gastrectomies at a high-volume cancer center in the United States. The study analyzed data for all patients undergoing curative gastrectomy for gastric adenocarcinoma from January 2007 to June 2017. Postoperative complications and disease-specific survival (DSS) were compared between surgical approaches. The median follow-up period for the 845 patients in this study was 38.5 months. The stage-stratified 5-year DSS did not differ significantly between open surgery (n = 534) and MIS (n = 311). The MIS approach resulted in significantly fewer complications, as confirmed by adjusted comparison (odds ratio [OR], 0.70; range, 0.49–1.00; p = 0.049). After adjustment, the two groups did not differ in terms of DSS (hazard ratio [HR], 0.83; range, 0.55–1.25; p = 0.362). The robotic operations (n = 190) had fewer conversions to open procedure (p = 0.010), a shorter operative time (212 vs 240 min; p < 0.001), more dissected nodes (27 vs 22; p < 0.001), fewer Clavien-Dindo grade ≥3 complications (5.8% vs 13.2%; p = 0.023), and a shorter postoperative stay (5 vs 6 days; p = 0.045) than the laparoscopic operations (n = 121). The DSS rate did not differ between the laparoscopic and robotic groups. The study findings demonstrated the long-term survival and oncologic equivalency of MIS gastrectomy and the open approach in a Western cohort, supporting the use of MIS at centers that have adequate experience with appropriately selected patients.
- Published
- 2021
- Full Text
- View/download PDF
32. Outcomes of Neoadjuvant Chemotherapy for Clinical Stages 2 and 3 Gastric Cancer Patients: Analysis of Timing and Site of Recurrence
- Author
-
Steven Brad Maron, David H. Ilson, Yelena Y. Janjigian, Laura H. Tang, Sam S. Yoon, Masaya Nakauchi, Daniel G. Coit, Mithat Gonen, Geoffrey Y. Ku, Murray F. Brennan, Vivian E. Strong, and Elvira L. Vos
- Subjects
medicine.medical_specialty ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Standard treatment ,Cancer ,Odds ratio ,medicine.disease ,Confidence interval ,Surgery ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Surgical oncology ,030220 oncology & carcinogenesis ,Medicine ,T-stage ,030211 gastroenterology & hepatology ,Stage (cooking) ,business - Abstract
This study aimed to analyze timing and sites of recurrence for patients receiving neoadjuvant chemotherapy for gastric cancer. Neoadjuvant chemotherapy followed by surgical resection is the standard treatment for locally advanced gastric cancer in the West, but limited information exists as to timing and patterns of recurrence in this setting. Patients with clinical stage 2 or 3 gastric cancer treated with neoadjuvant chemotherapy followed by curative-intent resection between January 2000 and December 2015 were analyzed for 5-year recurrence-free survival (RFS) as well as timing and site of recurrence. Among 312 identified patients, 121 (38.8%) experienced recurrence during a median follow-up period of 46 months. The overall 5-year RFS rate was 58.9%, with RFS rates of 95.8% for ypT0N0, 81% for ypStage 1, 77.4% for ypStage 2, and 22.9% for ypStage 3. The first site of recurrence was peritoneal for 49.6%, distant (not peritoneal) for 45.5%, and locoregional for 11.6% of the patients. The majority of the recurrences (84.3%) occurred within 2 years. Multivariate analysis showed that ypT4 status was an independent predictor for recurrence within 1 year after surgery (odds ratio, 2.58; 95% confidence interval, 1.10–6.08; p = 0.030). The majority of the recurrences for patients with clinical stage 2 or 3 gastric cancer who received neoadjuvant chemotherapy and underwent curative resection occurred within 2 years. After neoadjuvant chemotherapy, pathologic T stage was a useful risk predictor for early recurrence.
- Published
- 2021
- Full Text
- View/download PDF
33. Circulating tumor and invasive cell expression profiling predicts effective therapy in pancreatic cancer
- Author
-
Kenneth H. Yu, Jennifer Park, Avni Mittal, Ghassan K. Abou‐Alfa, Imane El Dika, Andrew S. Epstein, David H. Ilson, David P. Kelsen, Geoffrey Y. Ku, Jia Li, Wungki Park, Anna M. Varghese, Joanne F‐L Chou, Marinela Capanu, Brandon Cooper, Andrew Bartlett, Devan McCarthy, Vineet Sangar, Brian McCarthy, and Eileen M. O'Reilly
- Subjects
Pancreatic Neoplasms ,Cancer Research ,Oncology ,Paclitaxel ,Albumins ,Antineoplastic Combined Chemotherapy Protocols ,Leucovorin ,Humans ,Fluorouracil ,Prospective Studies ,Adenocarcinoma ,Deoxycytidine - Abstract
Pancreatic adenocarcinoma (PDAC) remains a refractory disease; however, modern cytotoxic chemotherapeutics can induce tumor regression and extend life. A blood-based, pharmacogenomic, chemosensitivity assay using gene expression profiling of circulating tumor and invasive cells (CTICs) to predict treatment response was previously developed. The combination regimen of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and gemcitabine/nab-paclitaxel (G/nab-P) are established frontline approaches for treating advanced PDAC; however, there are no validated biomarkers for treatment selection. A similar unmet need exists for choosing second-line therapy.The chemosensitivity assay was evaluated in metastatic PDAC patients presenting for frontline treatment. A prospective study enrolled patients (n = 70) before receiving either FOLFIRINOX or G/nab-P at a 1:1 ratio. Six milliliters of peripheral blood was collected at baseline and at time of disease progression. CTICs were isolated, gene-expression profiling was performed, and the assay was used to predict effective and ineffective chemotherapeutic agents. Treating physicians were blinded to the assay prediction results.Patients receiving an effective regimen as predicted by the chemosensitivity assay experienced significantly longer median progression-free survival (mPFS; 7.8 months vs. 4.2 months; hazard ratio [HR], 0.35; p = .0002) and median overall survival (mOS; 21.0 months vs. 9.7 months; HR, 0.40; p = .005), compared with an ineffective regimen. Assay prediction for effective second-line therapy was explored. The entire study cohort experienced favorable outcomes compared with historical controls, 7.1-month mPFS and 12.3-month mOS.Chemosensitivity assay profiling is a promising tool for guiding therapy in advanced PDAC. Further prospective validation is under way (clinicaltrials.gov NCT03033927).
- Published
- 2022
34. Clinical Importance of Clonal Hematopoiesis in Metastatic Gastrointestinal Tract Cancers
- Author
-
Bill H. Diplas, Ryan Ptashkin, Joanne F. Chou, Shalom Sabwa, Michael B. Foote, Benoit Rousseau, Guillem Argilés, James Robert White, Caitlin M. Stewart, Kelly Bolton, Sree B. Chalasani, Avni M. Desai, Zoe Goldberg, Ping Gu, Jia Li, Marina Shcherba, Alice Zervoudakis, Andrea Cercek, Rona Yaeger, Neil H. Segal, David H. Ilson, Geoffrey Y. Ku, Ahmet Zehir, Marinela Capanu, Yelena Y. Janjigian, Luis A. Diaz, and Steven B. Maron
- Subjects
General Medicine - Abstract
ImportanceClonal hematopoiesis (CH) has been associated with development of atherosclerosis and leukemia and worse survival among patients with cancer; however, the association with cancer therapy efficacy, in particular immune checkpoint blockade (ICB), and toxicity has not yet been established. Given the widespread use of ICB and the critical role hematopoietic stem cell–derived lymphocytes play in mediating antitumor responses, CH may be associated with therapeutic efficacy and hematologic toxicity.ObjectiveTo determine the association between CH and outcomes, hematologic toxicity, and therapeutic efficacy in patients with metastatic gastrointestinal tract cancers being treated with systemic therapy, both in the first-line metastatic treatment setting and in ICB.Design, Setting, and ParticipantsThis retrospective cohort study included 633 patients with stage IV colorectal (CRC) and esophagogastric (EGC) cancer who were treated with first-line chemotherapy and/or ICB at Memorial Sloan Kettering Cancer Center. Patients underwent matched tumor and peripheral blood DNA sequencing using the Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets next-generation sequencing assay between January 1, 2006, and December 31, 2020.ExposuresClonal hematopoiesis–related genetic alterations were identified by next-generation sequencing of patients’ tumor and normal blood buffy coat samples, with a subset of these CH alterations annotated as likely putative drivers (CH-PD) based upon previously established criteria.Main Outcomes and MeasuresPatients with CH and CH-PD in peripheral blood samples were identified, and these findings were correlated with survival outcomes (progression-free survival [PFS] and overall survival [OS]) during first-line chemotherapy and ICB, as well as baseline white blood cell levels and the need for granulocyte colony-stimulating factor (G-CSF) support.ResultsAmong the 633 patients included in the study (390 men [61.6%]; median age, 58 [IQR, 48-66] years), the median age was 52 (IQR, 45-63) years in the CRC group and 61 (IQR, 53-69) years in the EGC group. In the CRC group, 161 of 301 patients (53.5%) were men, compared with 229 of 332 patients (69.0%) in the EGC group. Overall, 62 patients (9.8%) were Asian, 45 (7.1%) were Black or African American, 482 (76.1%) were White, and 44 (7.0%) were of unknown race or ethnicity. Presence of CH was identified in 115 patients with EGC (34.6%) and 83 with CRC (27.6%), with approximately half of these patients harboring CH-PD (CRC group, 44 of 83 [53.0%]; EGC group, 55 of 115 [47.8%]). Patients with EGC and CH-PD exhibited a significantly worse median OS of 16.0 (95% CI, 11.6-22.3) months compared with 21.6 (95% CI, 19.6-24.3) months for those without CH-PD (P = .01). For patients with CRC and EGC, CH and CH-PD were not associated with PFS differences in patients undergoing ICB or first-line chemotherapy. Neither CH nor CH-PD were correlated with baseline leukocyte levels or increased need for G-CSF support.Conclusions and RelevanceThese findings suggest CH and CH-PD are not directly associated with the treatment course of patients with metastatic gastrointestinal tract cancer receiving cancer-directed therapy.
- Published
- 2023
- Full Text
- View/download PDF
35. Phase I/Ib study of crenolanib with ramucirumab and paclitaxel as second-line therapy for advanced esophagogastric adenocarcinoma
- Author
-
Ryan H, Moy, Megan, Greally, Joanne F, Chou, Jia, Li, Avni M, Desai, Sree B, Chalasani, Elizabeth, Won, David P, Kelsen, David H, Ilson, Yelena Y, Janjigian, Marinela, Capanu, and Geoffrey Y, Ku
- Subjects
Adult ,Male ,Dose-Response Relationship, Drug ,Esophageal Neoplasms ,Maximum Tolerated Dose ,Paclitaxel ,Adenocarcinoma ,Middle Aged ,Antibodies, Monoclonal, Humanized ,Progression-Free Survival ,Piperidines ,Stomach Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Benzimidazoles ,Female ,Aged - Abstract
Paclitaxel plus ramucirumab is a standard second-line regimen for patients with advanced gastric adenocarcinoma, but clinical benefit remains modest. One potential resistance mechanism to VEGFR2 inhibition is activation of the PDGF/PDGFR pathway, which can be blocked by the selective inhibitor crenolanib. Therefore, we performed a phase I/Ib study of crenolanib in combination with paclitaxel/ramucirumab.Patients with metastatic esophagogastric adenocarcinoma refractory to first-line therapy received escalating doses of crenolanib [60 mg twice daily (BID) to 100 mg three times daily (TID)] in combination with paclitaxel 80 mg/mWe enrolled 19 patients in the dose escalation phase and 8 patients in the dose expansion phase at the MTD of crenolanib 100 mg BID. Common grade 3/4 treatment-emergent adverse events included leukopenia (19%), anemia (11%) and neutropenia (11%). In the 14 patients treated at the MTD, 6-month PFS was 43% [95% confidence interval (CI) 23-78%] and the objective response rate (ORR) was 42% (95% CI 15-72%). The trial was terminated early due to withdrawal of crenolanib by the sponsor.The addition of crenolanib to paclitaxel/ramucirumab is safe and well-tolerated at a dose level up to 100 mg BID.NCT03193918. June 19, 2017.
- Published
- 2021
36. Approach to Resectable Gastric Cancer: Evolving Paradigm of Neoadjuvant and Adjuvant Treatment
- Author
-
Melissa A, Lumish and Geoffrey Y, Ku
- Subjects
Chemotherapy, Adjuvant ,Stomach Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Chemoradiotherapy, Adjuvant ,Adenocarcinoma ,Neoadjuvant Therapy - Abstract
Recent therapeutic advances have prolonged survival in patients with metastatic gastric cancer, though the prognosis for patients with locally advanced resectable gastric cancer remains poor. Long-term survival after resection of locally advanced gastric adenocarcinoma is dependent on early eradication of micrometastatic disease and optimal surgical resection. Preoperative therapy with a docetaxel-containing three-drug regimen has recently been shown to be superior to an anthracycline-containing three-drug regimen or two-drug therapy with a fluoropyrimidine and platinum. Chemoradiation is not essential and is reserved for patients with suboptimal resection. Emerging research strategies include introduction of pre- and postoperative checkpoint blockade and biomarker-directed therapy.
- Published
- 2021
37. Safety and Efficacy of Durvalumab and Tremelimumab Alone or in Combination in Patients with Advanced Gastric and Gastroesophageal Junction Adenocarcinoma
- Author
-
Yee Chao, Siddharth Sheth, Judson M. Englert, Yung-Jue Bang, Michael K. Gibson, Cheng Ean Chee, Ronan J. Kelly, Hyun Cheol Chung, Zev A. Wainberg, Jeeyun Lee, Peng He, Johanna C. Bendell, Rom Leidner, Philip Z Brohawn, Jennifer McDevitt, Mariela Blum-Murphy, Keun Wook Lee, Takeshi Omori, Heinz-Josef Lenz, Geoffrey Y. Ku, Crystal S. Denlinger, Daniel V.T. Catenacci, Marlon Rebelatto, and Khaldoun Almhanna
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Durvalumab ,Gastroesophageal Junction Adenocarcinoma ,Gastroenterology ,B7-H1 Antigen ,Circulating Tumor DNA ,law.invention ,0302 clinical medicine ,Randomized controlled trial ,law ,Antineoplastic Combined Chemotherapy Protocols ,Monoclonal ,80 and over ,Medicine ,CTLA-4 Antigen ,Prospective Studies ,Prospective cohort study ,Humanized ,6.2 Cellular and gene therapies ,Cancer ,Aged, 80 and over ,education.field_of_study ,Antibodies, Monoclonal ,Middle Aged ,Oncology ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Female ,Esophagogastric Junction ,Patient Safety ,medicine.drug ,Adult ,medicine.medical_specialty ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Population ,Adenocarcinoma ,Antibodies, Monoclonal, Humanized ,Article ,Antibodies ,Interferon-gamma ,Young Adult ,03 medical and health sciences ,Rare Diseases ,Stomach Neoplasms ,Clinical Research ,Internal medicine ,Humans ,Oncology & Carcinogenesis ,education ,Adverse effect ,Aged ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,030104 developmental biology ,Transcriptome ,Digestive Diseases ,business ,Tremelimumab - Abstract
Purpose: This randomized, multicenter, open-label, phase Ib/II study assessed durvalumab and tremelimumab in combination or as monotherapy for chemotherapy-refractory gastric cancer or gastroesophageal junction (GEJ) cancer. Patients and Methods: Second-line patients were randomized 2:2:1 to receive durvalumab plus tremelimumab (arm A), or durvalumab (arm B) or tremelimumab monotherapy (arm C), and third-line patients received durvalumab plus tremelimumab (arm D). A tumor-based IFNγ gene signature was prospectively evaluated as a potential predictive biomarker in second- and third-line patients receiving the combination (arm E). The coprimary endpoints were objective response rate and progression-free survival (PFS) rate at 6 months. Results: A total of 113 patients were treated: 6 in phase Ib and 107 (arm A, 27; arm B, 24; arm C, 12; arm D, 25; arm E, 19) in phase II. Overall response rates were 7.4%, 0%, 8.3%, 4.0%, and 15.8% in the five arms, respectively. PFS rates at 6 months were 6.1%, 0%, 20%, 15%, and 0%, and 12-month overall survival rates were 37.0%, 4.6%, 22.9%, 38.8%, and NA, respectively. Treatment-related grade 3/4 adverse events were reported in 17%, 4%, 42%, 16%, and 11% of patients, respectively. Conclusions: Response rates were low regardless of monotherapy or combination strategies. No new safety signals were identified. Including use of a tumor-based IFNγ signature and change in baseline and on-treatment circulating tumor DNA are clinically feasible and may be novel strategies to improve treatment response in this difficult-to-treat population.
- Published
- 2020
- Full Text
- View/download PDF
38. Clinical and Molecular Predictors of Response to Immune Checkpoint Inhibitors in Patients with Advanced Esophagogastric Cancer
- Author
-
Megan Greally, Luis A. Diaz, David B. Solit, Yaelle Tuvy, Laura H. Tang, David P. Kelsen, David H. Ilson, Geoffrey Y. Ku, Michael F. Berger, Jaclyn F. Hechtman, Marinela Capanu, Foysal Daian, Ritika Kundra, Nikolaus Schultz, Joanne F. Chou, Walid K. Chatila, Yelena Y. Janjigian, Marc Ladanyi, and Matthew Margolis
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Esophageal Neoplasms ,DNA Mutational Analysis ,Programmed Cell Death 1 Receptor ,Antibiotics ,ECOG Performance Status ,Disease ,B7-H1 Antigen ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,CTLA-4 Antigen ,Aged, 80 and over ,biology ,Middle Aged ,Prognosis ,Progression-Free Survival ,030220 oncology & carcinogenesis ,Female ,Esophagogastric Junction ,Antibody ,Adult ,medicine.medical_specialty ,medicine.drug_class ,Article ,Young Adult ,03 medical and health sciences ,Text mining ,Stomach Neoplasms ,Esophagogastric cancer ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,In patient ,Response Evaluation Criteria in Solid Tumors ,Aged ,Retrospective Studies ,business.industry ,Cancer ,medicine.disease ,030104 developmental biology ,Mutation ,biology.protein ,business ,Follow-Up Studies - Abstract
Purpose: Immune checkpoint inhibitors (ICI) are effective in only a minority of patients with esophagogastric cancer (EGC). Here, we aimed to identify predictors of durable clinical benefit to ICI in EGC. Experimental Design: Patients with advanced EGC treated with ICIs at Memorial Sloan Kettering Cancer Center (New York, NY) were identified. Clinicopathologic variables were assessed. In patients profiled by MSK-IMPACT–targeted sequencing, outcomes were correlated with tumor genomic features. Results: One-hundred sixty-one patients were treated with ICIs (110 with anti–PD-1/PD-L1 antibodies and 51 with anti–CTLA-4 and PD-1/PD-L1 antibodies). The median progression-free survival (PFS) and overall survival (OS) were 1.7 and 4.9 months, respectively. Greater number of disease sites (≥3), liver metastases, treatment with ≥3 prior therapies and ECOG performance status ≥2 were associated with poorer PFS and OS. Patients treated with combination ICI and those with PD-L1–positive tumors had improved outcomes. There was no difference in outcomes between patients treated with antibiotics during or in the 2 months preceding ICI treatment versus those who were not. Occurrence of irAEs was associated with improved OS. In genomically profiled tumors (n = 89), survival was associated with increasing tumor mutation burden (TMB). However, in multivariable analyses and when microsatellite unstable (MSI) patients were excluded, a significant association was no longer observed. Conclusions: In patients with advanced EGC, heavily pretreated patients, those with high-volume disease and/or poor PS were less likely to benefit from ICI. irAEs were associated with improved OS. TMB correlated with improved survival, but this association was not observed when MSI-high patients were excluded.
- Published
- 2019
- Full Text
- View/download PDF
39. Adjuvant chemotherapy for poor pathologic response after pre-operative chemoradiation in esophageal cancer: infeasible and illogical
- Author
-
Megan Greally and Geoffrey Y. Ku
- Subjects
Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Chemotherapy ,business.industry ,Adjuvant chemotherapy ,medicine.medical_treatment ,MEDLINE ,Esophageal cancer ,medicine.disease ,Pre operative ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,Carcinoma ,Pathologic Response ,Multimodal treatment ,business - Abstract
While recent decades have seen incremental improvements in the treatment of esophageal carcinoma, outcomes remain modest. The five-year overall survival (OS) rate for locally advanced esophageal cancer with surgery alone is 23–33% in contemporary studies (1-3). In locally advanced esophageal cancer, the risk of incomplete (R1) resection, local recurrence and systemic dissemination is significant. Numerous studies have demonstrated that the addition of chemotherapy and/or radiation to surgery improve outcomes, leading to multimodal treatment becoming standard-of-care. In particular, pre-operative chemoradiation has emerged as a standard-of-care in the US and Western Europe (4).
- Published
- 2019
- Full Text
- View/download PDF
40. Efficacy of Combined VEGFR1-3, PDGFα/β, and FGFR1-3 Blockade Using Nintedanib for Esophagogastric Cancer
- Author
-
Nikolaus Schultz, Yaelle Tuvy, Zoe Goldberg, Michael F. Berger, Geoffrey Y. Ku, Marinela Capanu, Laura H. Tang, Sree Bhavani Chalasani, Joanne F. Chou, Jaclyn F. Hechtman, Mark A. Schattner, Elizabeth Won, Michelle S. Boyar, David P. Kelsen, Walid K. Chatila, David H. Ilson, Azfar Basunia, David B. Solit, Avni M. Desai, and Yelena Y. Janjigian
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Indoles ,Esophageal Neoplasms ,medicine.medical_treatment ,Antineoplastic Agents ,Disease ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Stomach Neoplasms ,Internal medicine ,Biomarkers, Tumor ,medicine ,Clinical endpoint ,Humans ,Molecular Targeted Therapy ,Protein Kinase Inhibitors ,Aged ,Chemotherapy ,business.industry ,Computational Biology ,Receptor Protein-Tyrosine Kinases ,Middle Aged ,medicine.disease ,Confidence interval ,Blockade ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Adenocarcinoma ,Female ,Nintedanib ,business ,Signal Transduction - Abstract
Purpose: VEGFR2-directed therapy is commonly used to treat metastatic esophagogastric cancer, but disease progresses in most patients within months. Therapeutic resistance is likely mediated in part by co-occurring amplifications of the genes for multiple oncogenic receptor tyrosine kinases (RTK). We therefore tested the efficacy of combined inhibition of VEGFR1-3, PDGFα/β, and FGFR1-3 using nintedanib. Patients and Methods: Patients with metastatic esophagogastric adenocarcinoma and disease progression on first-line chemotherapy were treated with nintedanib 200 mg twice daily. The primary endpoint was progression-free survival (PFS) at 6 months; secondary endpoints included tumor response and safety. Tumor biopsies were profiled by targeted capture next-generation sequencing (NGS) to identify molecular predictors of drug response. Results: The study achieved its primary endpoint; 6 of 32 patients (19%) were progression-free at 6 months. With a median follow-up of 14.5 months among survivors, median overall survival (OS) was 14.2 months [95% confidence interval (CI), 10.8 months–NR]. Nintedanib was well tolerated; grade ≥ 3 toxicities were uncommon and included grade 3 hypertension (15%) and liver enzyme elevation (4%). FGFR2 alterations were identified in 18% of patients but were not predictive of clinical outcome on nintedanib therapy. Alterations in cell-cycle pathway genes were associated with worse median PFS (1.61 months for patients with cell-cycle pathway alterations vs. 2.66 months for patients without, P = 0.019). Conclusions: Nintedanib treatment resulted in modest disease stabilization in patients with metastatic esophagogastric cancer. Alterations in cell-cycle pathway genes and increased global copy-number alteration (CNA) burden warrant further study as prognostic or predictive biomarkers.
- Published
- 2019
- Full Text
- View/download PDF
41. Positron-Emission Tomography Scan–Directed Chemoradiation for Esophageal Squamous Cell Carcinoma: No Benefit for a Change in Chemotherapy in Positron-Emission Tomography Nonresponders
- Author
-
Bernard J. Park, Abraham J. Wu, David P. Kelsen, Daniela Molena, Valerie W. Rusch, Geoffrey Y. Ku, Manjit S. Bains, David H. Ilson, Yelena Y. Janjigian, Megan Greally, Karyn A. Goodman, and Joanne F. Chou
- Subjects
Adult ,Male ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Esophageal Neoplasms ,medicine.medical_treatment ,Standardized uptake value ,Gastroesophageal Junction Adenocarcinoma ,Article ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Fluorodeoxyglucose F18 ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Medicine ,Young adult ,Survival rate ,Aged ,Retrospective Studies ,Aged, 80 and over ,Chemotherapy ,medicine.diagnostic_test ,business.industry ,Induction chemotherapy ,Retrospective cohort study ,Chemoradiotherapy ,Induction Chemotherapy ,Middle Aged ,Prognosis ,Survival Rate ,030104 developmental biology ,Oncology ,Positron emission tomography ,Positron-Emission Tomography ,030220 oncology & carcinogenesis ,Female ,Esophageal Squamous Cell Carcinoma ,Radiology ,Radiopharmaceuticals ,business ,Follow-Up Studies - Abstract
INTRODUCTION: Preoperative or definitive chemoradiation is an accepted treatment for locally advanced esophageal squamous cell carcinoma (ESCC). The MUNICON study showed that positron-emission tomography (PET) response following induction chemotherapy was predictive of outcomes in patients with gastroesophageal junction adenocarcinoma. We evaluated the predictive value of PET following induction chemotherapy in ESCC patients and assessed the impact of changing chemotherapy during radiation in PET nonresponders. METHODS: We retrospectively reviewed all patients with locally advanced ESCC who received induction chemotherapy and chemoradiation; all patients had a PET before and after induction chemotherapy. Survival was calculated from date of repeat PET using Kaplan-Meier analysis and compared between groups using the log-rank test. RESULTS: Of 111 patients, 70 (63%) were PET responders (defined as a 35% or more decrease in maximum standard uptake value) to induction chemotherapy. PET responders received the same chemotherapy during radiation. Of 41 PET nonresponders, 16 continued with the same chemotherapy and 25 were changed to alternative chemotherapy with radiation. Median progression-free survival (70.1 months versus 7.1 months, p < 0.01) and overall survival (84.8 months versus 17.2 months, p < 0.01) were improved for PET responders versus nonresponders. Median progression-free survival and overall survival for PET nonresponders who changed chemotherapy versus those who did not were 6.4 months versus 8.3 months (p = 0.556) and 14.1 versus 17.2 months (p = 0.81), respectively. CONCLUSIONS: PET after induction chemotherapy highly predicts for outcomes in ESCC patients who receive chemoradiation. However, our results suggest that PET nonresponders do not benefit from changing chemotherapy during radiation. Future trials should use PET nonresponse after induction chemotherapy to identify poor prognosis patients for novel therapies.
- Published
- 2019
- Full Text
- View/download PDF
42. Prognostic significance of T-cell-inflamed gene expression profile and PD-L1 expression in patients with esophageal cancer
- Author
-
Ting Wu, Morten Ladekarl, Hyun Cheol Chung, Hyunki Kim, Marie Louise Jespersen, Senaka Peter, Marianne Nordsmark, Torben Steiniche, Jeanette Bæhr Georgsen, Matthew J. Marton, Geoffrey Y. Ku, Laura H. Tang, David P. Kelsen, Hyo Song Kim, Carly Fein, and Sun Young Rha
- Subjects
Oncology ,Male ,squamous cell carcinoma ,Cancer Research ,medicine.medical_specialty ,Esophageal Neoplasms ,T-Lymphocytes ,retrospective study ,T‐lymphocytes ,B7-H1 Antigen ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,Radiology, Nuclear Medicine and imaging ,Stage (cooking) ,T-lymphocytes ,RC254-282 ,Research Articles ,Aged ,Retrospective Studies ,adenocarcinoma ,Performance status ,Proportional hazards model ,business.industry ,Gene Expression Profiling ,Hazard ratio ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Clinical Cancer Research ,Histology ,Retrospective cohort study ,Esophageal cancer ,medicine.disease ,Prognosis ,Adenocarcinoma ,Female ,business ,Research Article - Abstract
Purpose The ability of the T‐cell–inflamed gene expression profile (GEP) to predict clinical outcome in esophageal cancer (EC) is unknown. This retrospective observational study assessed the prognostic value of GEP and programmed death ligand 1 (PD‐L1) expression in patients with EC treated in routine clinical practice. Methods Tumor samples of 294 patients from three centers in Denmark, South Korea, and the United States, collected between 2005 and 2017, were included. T‐cell–inflamed GEP score was defined as non‐low or low using a cutoff of −1.54. A combined positive score (CPS) ≥10 was defined as PD‐L1 expression positivity. Associations between overall survival (OS) and GEP status and PD‐L1 expression were explored by Cox proportional hazards models adjusting for age, sex, histology, stage, and performance status. Results Median age was 65 years; 63% of patients had adenocarcinoma (AC) and 37% had squamous cell carcinoma (SCC). Thirty‐six percent of tumors were GEP non‐low, with higher prevalence in AC (46%) than SCC (18%). Twenty‐one percent were PD‐L1–positive: 32% in South Korean samples versus 16% in non‐Asian samples and 26% in SCC versus 18% in AC. GEP scores and PD‐L1 CPS were weakly correlated (Spearman’s R = 0.363). OS was not significantly associated with GEP status (non‐low vs low; adjusted hazard ratio, 0.91 [95% CI, 0.69–1.19]) or PD‐L1 expression status. Conclusion Neither GEP nor PD‐L1 expression was a prognostic marker in Asian and non‐Asian patients with EC., In a cohort of 294 patients with esophageal adenocarcinoma and esophageal squamous cell carcinoma from Denmark, South Korea, and the United States, T‐cell–inflamed gene expression profile (GEP) score (non‐low vs low) and PD‐L1 expression status (CPS ≥10 vs CPS
- Published
- 2021
- Full Text
- View/download PDF
43. Abstract CT017: Clinical and biomarker activity of PRS-343, a bispecific fusion protein targeting 4-1BB and HER2, from a phase 1 study in patients with advanced solid tumors (Study PRS-343-PCS_04_16)
- Author
-
Lynn Mar, Geoffrey Y. Ku, Sara A. Hurvitz, Markus Zettl, Patrick Jolicoeur, Paula R. Pohlmann, Corinna Schlosser, Johanna C. Bendell, Kayti Aviano, Rachna T. Shroff, Anuradha Krishnamurthy, Naimish Pandya, Aizea Morales Kastresana, Anthony W. Tolcher, Ingmar Bruns, Alice Bexon, Sarina Anne Piha-Paul, and Shane Olwill
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Internal medicine ,Medicine ,Biomarker (medicine) ,In patient ,business - Abstract
The authors did not submit an updated abstract. The original abstract should be considered final. Citation Format: Sarina Piha-Paul, Johanna C. Bendell, Anthony Tolcher, Rachna Shroff, Paula R. Pohlmann, Sara A. Hurvitz, Anuradha Krishnamurthy, Naimish Pandya, Shane A. Olwill, Markus Zettl, Kayti Aviano, Lynn Mar, Patrick Jolicoeur, Aizea Morales Kastresana, Corinna Schlosser, Ingmar Bruns, Alice Bexon, Geoffrey Y. Ku. Clinical and biomarker activity of PRS-343, a bispecific fusion protein targeting 4-1BB and HER2, from a phase 1 study in patients with advanced solid tumors (Study PRS-343-PCS_04_16) [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr CT017.
- Published
- 2021
- Full Text
- View/download PDF
44. Can 18F-FDG PET/CT Radiomics Features Predict Clinical Outcomes in Patients with Locally Advanced Esophageal Squamous Cell Carcinoma?
- Author
-
Vetri Sudar Jayaprakasam, Peter Gibbs, Natalie Gangai, Raazi Bajwa, Ramon E. Sosa, Randy Yeh, Megan Greally, Geoffrey Y. Ku, Marc J. Gollub, and Viktoriya Paroder
- Subjects
Cancer Research ,Oncology ,esophageal squamous cell carcinoma ,PET/CT ,radiomics ,progression-free survival ,overall survival - Abstract
This study aimed to assess the usefulness of radiomics features of 18F-FDG PET/CT in patients with locally advanced esophageal cancers (ESCC) in predicting outcomes such as clinical tumor (cT) and nodal (cN) categories, PET response to induction chemotherapy (PET response), progression-free survival (PFS), and overall survival (OS). Pretreatment PET/CT images from patients who underwent concurrent chemoradiotherapy from July 2002 to February 2017 were segmented, and data were split into training and test sets. Model development was performed on the training datasets and a maximum of five features were selected. Final diagnostic accuracies were determined using the test dataset. A total of 86 PET/CTs (58 men and 28 women, mean age 65 years) were segmented. Due to small lesion size, 12 patients were excluded. The diagnostic accuracies as derived from the CT, PET, and combined PET/CT test datasets were as follows: cT category—70.4%, 70.4%, and 81.5%, respectively; cN category—69.0%, 86.2%, and 86.2%, respectively; PET response—60.0%, 66.7%, and 70.0%, respectively; PFS—60.7%, 75.0%, and 75.0%, respectively; and OS—51.7%, 55.2%, and 62.1%, respectively. A radiomics assessment of locally advanced ESCC has the potential to predict various clinical outcomes. External validation of these models would be further helpful.
- Published
- 2022
- Full Text
- View/download PDF
45. 1380P Phase (Ph) II study of zanidatamab + chemotherapy (chemo) in first-line (1L) HER2 expressing gastroesophageal adenocarcinoma (GEA)
- Author
-
Geoffrey Y. Ku, R. Mehta, Sun Young Rha, C. S. Denlinger, T. Mwatha, Yeul Hong Kim, Jaffer A. Ajani, K-W. Lee, Y-K. Kang, Elena Elimova, D-Y. Oh, S. Iqbal, Y. M. Seol, and J. Grim
- Subjects
Chemotherapy ,Gastroesophageal adenocarcinoma ,Oncology ,business.industry ,Phase (matter) ,First line ,medicine.medical_treatment ,Cancer research ,Medicine ,Hematology ,business - Published
- 2021
- Full Text
- View/download PDF
46. 1421P EGFR inhibition in EGFR-amplified esophagogastric cancer (EGC): Retrospective global experience
- Author
-
Pashtoon Murtaza Kasi, S. Moya, Steven Brad Maron, Joseph Chao, J.W. Lee, Filippo Pietrantonio, Samuel J. Klempner, Zev A. Wainberg, Yoshiaki Nakamura, Daniel Virgil Thomas Catenacci, Geoffrey Y. Ku, Kohei Shitara, Shumei Kato, Russell D. Petty, M.J. Emmett, N. Uboha, U. Disel, S. Chalasani, and Federica Morano
- Subjects
Oncology ,medicine.medical_specialty ,Esophagogastric cancer ,business.industry ,Egfr inhibition ,Internal medicine ,Medicine ,Hematology ,business - Published
- 2021
- Full Text
- View/download PDF
47. Prognosis after neoadjuvant chemoradiation or chemotherapy for locally advanced gastro-oesophageal junctional adenocarcinoma
- Author
-
Daniel G. Coit, T Nobel, Rebecca Carr, David R. Jones, Manjit S. Bains, A Russo, Geoffrey Y. Ku, Kay See Tan, D. Ilson, Laura H. Tang, Daniela Molena, Arianna Barbetta, Abraham J. Wu, Yelena Y. Janjigian, Vivian E. Strong, Elvira L. Vos, Sam S. Yoon, Meier Hsu, and Masaya Nakauchi
- Subjects
Male ,medicine.medical_specialty ,Esophageal Neoplasms ,medicine.medical_treatment ,New York ,Antineoplastic Agents ,Adenocarcinoma ,Gastroenterology ,Article ,Postoperative Complications ,Internal medicine ,medicine ,Humans ,Cumulative incidence ,Lymph node ,Aged ,Neoplasm Staging ,Retrospective Studies ,Chemotherapy ,business.industry ,Incidence ,Hazard ratio ,Cancer ,Chemoradiotherapy, Adjuvant ,Middle Aged ,medicine.disease ,Prognosis ,Log-rank test ,Esophagectomy ,Survival Rate ,Exact test ,medicine.anatomical_structure ,Surgery ,Female ,Esophagogastric Junction ,business ,Follow-Up Studies - Abstract
Trials typically group cancers of the gastro-oesophageal junction (GOJ) with oesophageal or gastric cancer when studying neoadjuvant chemoradiation and perioperative chemotherapy, so the results may not be fully applicable to GOJ cancer. Because optimal neoadjuvant treatment for GOJ cancer remains controversial, outcomes with neoadjuvant chemoradiation versus chemotherapy for locally advanced GOJ adenocarcinoma were compared retrospectively.Data were collected from all patients who underwent neoadjuvant treatment followed by surgery for adenocarcinoma located at the GOJ at a single high-volume institution between 2002 and 2017. Postoperative major complications and mortality were compared between groups using Fisher's exact test. Overall survival (OS) and disease-free survival (DFS) were assessed by log rank test and multivariable Cox regression analyses. Cumulative incidence functions were used to estimate recurrence, and groups were compared using Gray's test.Of 775 patients, 650 had neoadjuvant chemoradiation and 125 had chemotherapy. These groups were comparable in terms of clinical tumour and lymph node categories, although the chemoradiation group had greater proportions of white men, complete pathological response to chemotherapy, and smaller proportions of diffuse cancer, poor differentiation, and neurovascular invasion. Postoperative major complications (20.0 versus 17.6 per cent) and 30-day mortality (1.7 versus 1.6 per cent) were not significantly different between the chemoradiation and chemotherapy groups. After adjustment, type of therapy (chemoradiation versus chemotherapy) was not significantly associated with OS (hazard ratio (HR) 1.26, 95 per cent c.i. 0.96 to 1.67) or DFS (HR 1.27, 0.98 to 1.64). Type of recurrence (local, regional, or distant) did not differ after neoadjuvant chemoradiation versus chemotherapy.In patients undergoing surgical resection for locally advanced adenocarcinoma of the GOJ, OS and DFS did not differ significantly between patients who had neoadjuvant chemoradiation compared with chemotherapy.Treating advanced cancer of the gastro-oesophageal junction (GOJ) poses a challenge given its location in the distal oesophagus and proximal stomach, and whether it should be treated as oesophageal or gastric cancer. Given the indistinct location, it is unclear whether GOJ cancer should be treated with neoadjuvant chemoradiation, which is the treatment of choice for advanced oesophageal cancers, or perioperative chemotherapy, which is the treatment of choice for advanced gastric cancers. Few studies have addressed treatment options specifically for GOJ cancers. This study investigated whether there was a difference in survival between patients with GOJ cancer who were treated with chemoradiation versus chemotherapy.
- Published
- 2021
48. Phase II Single Arm Study of Durvalumab and Tremelimumab with Concurrent Radiotherapy in Patients with Mismatch Repair Proficient Metastatic Colorectal Cancer
- Author
-
Joanne F. Chou, Anna M. Varghese, Pallavi Vedantam, Andrea Cercek, Andreas Rimner, Karuna Ganesh, Travis J. Hollmann, Nancy E. Kemeny, Joseph P. Erinjeri, Yoshiya Yamada, Paul B. Romesser, Diane Reidy-Lagunes, Efsevia Vakiani, Aliya Holland, Neil H. Segal, T. Jonathan Yang, Geoffrey Y. Ku, Abraham J. Wu, Mark L. Solter, Martinique Ogle, Martin R. Weiser, Kathleen C. McAuliffe, Christopher H. Crane, Phillip Wong, Stephen B. Solomon, Danny N. Khalil, John J. Cuaron, Louise Catherine Connell, Marinela Capanu, Krishna Juluru, Taha Merghoub, Leonard B. Saltz, Zsofia K. Stadler, Rona Yaeger, Pamela Vaiskauskas, Ghassan K. Abou-Alfa, David Faleck, and Matthew Adamow
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Durvalumab ,Colorectal cancer ,medicine.medical_treatment ,Phases of clinical research ,CD8-Positive T-Lymphocytes ,Antibodies, Monoclonal, Humanized ,DNA Mismatch Repair ,Article ,03 medical and health sciences ,0302 clinical medicine ,Median follow-up ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Immune Checkpoint Inhibitors ,Response Evaluation Criteria in Solid Tumors ,Aged ,business.industry ,Antibodies, Monoclonal ,Immunotherapy ,Chemoradiotherapy ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Radiation therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Feasibility Studies ,Female ,business ,Colorectal Neoplasms ,Tremelimumab ,medicine.drug ,Follow-Up Studies - Abstract
Purpose:Immune checkpoint inhibition (ICI) alone is not active in mismatch repair–proficient (MMR-P) metastatic colorectal cancer (mCRC), nor does radiotherapy alone result in objective systemic benefit. However, combined radiotherapy plus ICI can induce systemic antitumor immunity in preclinical and clinical models.Patients and Methods:In this single-center, phase II study, patients with chemotherapy-refractory MMR-P mCRC received durvalumab 1,500 mg plus tremelimumab 75 mg every 4 weeks plus radiotherapy. The primary endpoint was objective response rate (ORR) in nonirradiated lesions. Treatment and efficacy were correlated with peripheral immune cell profiles.Results:We enrolled 24 patients, and report outcomes after a median follow-up of 21.8 (range: 15.9–26.3) months. The ORR was 8.3% (2 patients) [95% confidence interval (CI), 1.0–27.0]. The median progression-free survival was 1.8 (95% CI, 1.7–1.9) months, median overall survival was 11.4 (95% CI, 10.1–17.4) months. Twenty five percent of patients (n = 6) had treatment-related grade 3–4 adverse events. We observed increased circulating CD8+ T lymphocyte activation, differentiation, and proliferation in patients with objective response.Conclusions:This combination of radiotherapy plus ICI study did not meet the prespecified endpoint criteria to be considered worthwhile for further study. However, rare instances of systemic immune augmentation and regression in nonirradiated lesions were observed (an abscopal response). Combination durvalumab and tremelimumab plus radiotherapy is feasible in MMR-P mCRC with a manageable safety profile. Further studies of novel immunotherapy combinations, and identification of biomarkers predictive of abscopal response are warranted.
- Published
- 2021
49. Association of Obesity with Worse Operative and Oncologic Outcomes for Patients Undergoing Gastric Cancer Resection
- Author
-
Steven Brad Maron, Elvira L. Vos, Vivian E. Strong, Murray F. Brennan, David H. Ilson, Daniel G. Coit, Geoffrey Y. Ku, Yelena Y. Janjigian, Sam S. Yoon, Laura H. Tang, Masaya Nakauchi, and Mithat Gonen
- Subjects
medicine.medical_specialty ,medicine.medical_treatment ,Gastroenterology ,Article ,Body Mass Index ,03 medical and health sciences ,0302 clinical medicine ,Surgical oncology ,Gastrectomy ,Stomach Neoplasms ,Internal medicine ,medicine ,Humans ,Obesity ,Retrospective Studies ,Chemotherapy ,business.industry ,Hazard ratio ,Cancer ,medicine.disease ,Confidence interval ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Cohort ,030211 gastroenterology & hepatology ,Surgery ,business ,Body mass index - Abstract
BACKGROUND. How obesity has an impact on operative and oncologic outcomes for gastric cancer patients is unclear, and the influence of obesity on response to neoadjuvant chemotherapy (NAC) has not been evaluated. METHODS. Patients who underwent curative gastrectomy for primary gastric cancer between 2000 and 2018 were retrospectively identified. After stratification for NAC, operative morbidity, mortality, overall survival (OS), and disease-specific survival (DSS) were compared among three body mass index (BMI) categories: normal BMI ( In the upfront surgery cohort, according to the BMI, OS and DSS did not differ, whereas in the NAC cohort, severe obesity was independently associated with worse OS (hazard ratio [HR], 1.87; 95 % confidence interval [CI], 1.01–3.48; p = 0.047) and disease-specific survival [DSS] (HR, 2.08; 95 % CI, 1.07–4.05; p = 0.031). CONCLUSION. For the gastric cancer patients undergoing curative gastrectomy, obesity was associated with significantly lower rates of pathologic response to NAC and more postoperative complications, as well as shorter OS and DSS for the patients receiving NAC.
- Published
- 2021
50. Next-Generation Sequencing of 487 Esophageal Adenocarcinomas Reveals Independently Prognostic Genomic Driver Alterations and Pathways
- Author
-
Abraham J. Wu, Sergio De La Torre, Jaclyn F. Hechtman, Yelena Y. Janjigian, David R. Jones, Laura H. Tang, Kay See Tan, Michael F. Berger, Daniela Molena, Assem Patel, Steven Brad Maron, Pari Shah, Samuel C Nussenzweig, David B. Solit, Smita Sihag, Geoffrey Y. Ku, Henry Walch, Francisco Sanchez-Vega, Walid K. Chatila, Nikolaus Schultz, and Meier Hsu
- Subjects
0301 basic medicine ,Genome instability ,Oncology ,Cancer Research ,medicine.medical_specialty ,Disease ,Adenocarcinoma ,medicine.disease_cause ,Article ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Trastuzumab ,Internal medicine ,medicine ,Biomarkers, Tumor ,Humans ,business.industry ,High-Throughput Nucleotide Sequencing ,Cell cycle ,medicine.disease ,Prognosis ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,Mutation ,KRAS ,business ,medicine.drug - Abstract
Purpose: To delineate recurrent oncogenic driver alterations and dysregulated pathways in esophageal adenocarcinoma and to assess their prognostic value. Experimental Design: We analyzed a large cohort of patients with lower esophageal and junctional adenocarcinoma, prospectively sequenced by MSK-IMPACT with high-quality clinical annotation. Patients were subdivided according to treatment intent, curative versus palliative, which closely mirrored clinical staging. Genomic features, alterations, and pathways were examined for association with overall survival using Cox proportional hazard models, adjusted for relevant clinicopathologic factors knowable at the time of diagnosis. Results: Analysis of 487 patients revealed 16 oncogenic driver alterations, mostly amplifications, present in ≥5% of patients. Patients in the palliative-intent cohort, compared with those in the curative-intent cohort, were more likely to have metastatic disease, ERBB2 amplifications, Cell-cycle and RTK–RAS pathway alterations, as well as a higher fraction of genome altered and rate of whole-genome doubling. In multivariable analyses, CDKN2A alterations, SMAD4 alterations, KRAS amplifications, Cell-cycle and TGFβ pathways, and overall number of oncogenic drivers were independently associated with worse overall survival. ERBB2 amplification was associated with improved survival, presumably due to trastuzumab therapy. Conclusions: Our study suggests that higher levels of genomic instability are associated with more advanced disease in esophageal adenocarcinoma. Furthermore, CDKN2A, KRAS, and SMAD4 represent prognostic biomarkers, given their strong association with poor survival.
- Published
- 2020
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.