Thanyanan Reungwetwattana
Mahidol University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Thanyanan Reungwetwattana.
The New England Journal of Medicine | 2018
Jean-Charles Soria; Yuichiro Ohe; Johan Vansteenkiste; Thanyanan Reungwetwattana; Busyamas Chewaskulyong; Ki Hyeong Lee; Arunee Dechaphunkul; Fumio Imamura; Naoyuki Nogami; Takayasu Kurata; Isamu Okamoto; Caicun Zhou; Byoung Chul Cho; Ying Cheng; Eun Kyung Cho; Pei Jye Voon; David Planchard; Wu-Chou Su; Jhanelle E. Gray; Siow-Ming Lee; Rachel Hodge; Marcelo Marotti; Yuri Rukazenkov; Suresh S. Ramalingam
Background Osimertinib is an oral, third‐generation, irreversible epidermal growth factor receptor tyrosine kinase inhibitor (EGFR‐TKI) that selectively inhibits both EGFR‐TKI–sensitizing and EGFR T790M resistance mutations. We compared osimertinib with standard EGFR‐TKIs in patients with previously untreated, EGFR mutation–positive advanced non–small‐cell lung cancer (NSCLC). Methods In this double‐blind, phase 3 trial, we randomly assigned 556 patients with previously untreated, EGFR mutation–positive (exon 19 deletion or L858R) advanced NSCLC in a 1:1 ratio to receive either osimertinib (at a dose of 80 mg once daily) or a standard EGFR‐TKI (gefitinib at a dose of 250 mg once daily or erlotinib at a dose of 150 mg once daily). The primary end point was investigator‐assessed progression‐free survival. Results The median progression‐free survival was significantly longer with osimertinib than with standard EGFR‐TKIs (18.9 months vs. 10.2 months; hazard ratio for disease progression or death, 0.46; 95% confidence interval [CI], 0.37 to 0.57; P<0.001). The objective response rate was similar in the two groups: 80% with osimertinib and 76% with standard EGFR‐TKIs (odds ratio, 1.27; 95% CI, 0.85 to 1.90; P=0.24). The median duration of response was 17.2 months (95% CI, 13.8 to 22.0) with osimertinib versus 8.5 months (95% CI, 7.3 to 9.8) with standard EGFR‐TKIs. Data on overall survival were immature at the interim analysis (25% maturity). The survival rate at 18 months was 83% (95% CI, 78 to 87) with osimertinib and 71% (95% CI, 65 to 76) with standard EGFR‐TKIs (hazard ratio for death, 0.63; 95% CI, 0.45 to 0.88; P=0.007 [nonsignificant in the interim analysis]). Adverse events of grade 3 or higher were less frequent with osimertinib than with standard EGFR‐TKIs (34% vs. 45%). Conclusions Osimertinib showed efficacy superior to that of standard EGFR‐TKIs in the first‐line treatment of EGFR mutation–positive advanced NSCLC, with a similar safety profile and lower rates of serious adverse events. (Funded by AstraZeneca; FLAURA ClinicalTrials.gov number, NCT02296125.)
Journal of Thoracic Oncology | 2016
Anne S. Tsao; Giorgio V. Scagliotti; Paul A. Bunn; David P. Carbone; Graham W. Warren; Chunxue Bai; Harry J. de Koning; A. Uraujh Yousaf-Khan; Annette McWilliams; Ming-Sound Tsao; Prasad S. Adusumilli; Ramón Rami-Porta; Hisao Asamura; Paul Van Schil; Gail Darling; Suresh S. Ramalingam; Daniel R. Gomez; Kenneth E. Rosenzweig; Stefan Zimmermann; Solange Peters; Sai-Hong Ignatius Ou; Thanyanan Reungwetwattana; Pasi A. Jänne; Tony Mok; Heather A. Wakelee; Robert Pirker; Julien Mazieres; Julie R. Brahmer; Yang Zhou; Roy S. Herbst
ABSTRACT Lung cancer continues to be a major global health problem; the disease is diagnosed in more than 1.6 million new patients each year. However, significant progress is underway in both the prevention and treatment of lung cancer. Lung cancer therapy has now emerged as a “role model” for precision cancer medicine, with several important therapeutic breakthroughs occurring during 2015. These advances have occurred primarily in the immunotherapy field and in treatments directed against tumors harboring specific oncogenic drivers. Our knowledge about molecular mechanisms for oncogene‐driven tumors and about resistance to targeted therapies has increased quickly over the past year. As a result, several regulatory approvals of new agents that significantly improve survival and quality of life for patients with lung cancer who have advanced disease have occurred. The International Association for the Study of Lung Cancer has gathered experts in different areas of lung cancer research and management to summarize the most significant scientific advancements related to prevention and therapy of lung cancer during the past year.
Journal of Clinical Oncology | 2017
Milind Javle; Maeve Aine Lowery; Rachna T. Shroff; Karl Heinz Weiss; Christoph Springfeld; Mitesh J. Borad; Ramesh K. Ramanathan; Lipika Goyal; Saeed Sadeghi; Teresa Macarulla; Anthony B. El-Khoueiry; Ivan Borbath; Su Pin Choo; Do Youn Oh; Philip A. Philip; Li-Tzong Chen; Thanyanan Reungwetwattana; Eric Van Cutsem; Kun-Huei Yeh; Kristen K. Ciombor; Richard S. Finn; Anuradha Patel; Suman Sen; Dale Porter; Randi Isaacs; Andrew X. Zhu; Ghassan K. Abou-Alfa; Tanios Bekaii-Saab
Purpose No standard treatment exists for patients with cholangiocarcinoma for whom first-line gemcitabine-based therapy fails. Fibroblast growth factor receptor 2 ( FGFR2) fusions/translocations are present in 13% to 17% of intrahepatic cholangiocarcinomas. BGJ398, an orally bioavailable, selective pan-FGFR kinase inhibitor, has shown preliminary clinical activity against tumors with FGFR alterations. Methods A multicenter, open-label, phase II study ( ClinicalTrials.gov identifier: NCT02150967) evaluated BGJ398 antitumor activity in patients age ≥ 18 years with advanced or metastatic cholangiocarcinoma containing FGFR2 fusions or other FGFR alterations whose disease had progressed while receiving prior therapy. Patients received BGJ398 125 mg once daily for 21 days, then 7 days off (28-day cycles). The primary end point was investigator-assessed overall response rate. Results Sixty-one patients (35 women; median age, 57 years) with FGFR2 fusion (n = 48), mutation (n = 8), or amplification (n = 3) participated. At the prespecified data cutoff (June 30, 2016), 50 patients had discontinued treatment. All responsive tumors contained FGFR2 fusions. The overall response rate was 14.8% (18.8% FGFR2 fusions only), disease control rate was 75.4% (83.3% FGFR2 fusions only), and estimated median progression-free survival was 5.8 months (95% CI, 4.3 to 7.6 months). Adverse events included hyperphosphatemia (72.1% all grade), fatigue (36.1%), stomatitis (29.5%), and alopecia (26.2%). Grade 3 or 4 treatment-related adverse events occurred in 25 patients (41%) and included hyperphosphatemia (16.4%), stomatitis (6.6%), and palmar-plantar erythrodysesthesia (4.9%). Conclusion BGJ398 is a first-in-class FGFR kinase inhibitor with manageable toxicities that shows meaningful clinical activity against chemotherapy-refractory cholangiocarcinoma containing FGFR2 fusions. This promising antitumor activity supports continued development of BGJ398 in this highly selected patient population.
Lung Cancer | 2014
Julian R. Molina; Nathan R. Foster; Thanyanan Reungwetwattana; Garth D. Nelson; Andrew V. Grainger; Preston D. Steen; Philip J. Stella; Randolph S. Marks; Jack C. Wright; Alex A. Adjei
INTRODUCTION To assess the efficacy and the Src-kinase inhibitor saracatinib (AZD-0530) after four cycles of platinum-based chemotherapy for extensive stage small cell lung cancer (SCLC). METHODS Patients with at least stable disease received saracatinib at a dose of 175 mg/day by mouth until disease progression, unacceptable toxicity, or patient refusal. The primary endpoint was the 12-week progression-free survival (PFS) rate from initiation of saracatinib treatment. Planned interim analysis in first 20 patients, where 13 or more patients alive and progression-free at 12-weeks would allow continued enrollment to 40 total patients. RESULTS All 23 evaluable patients received platinum based standard chemotherapy. Median age was 58 years (range: 48-82). 96% of patients had a performance status of 0/1. Median of two cycles given (range: 1-34). All 23 (100%) patients have ended treatment, most for disease progression (19/23). The 12-week PFS rate was 26% (6/23; 95% CI: 10-48%). From start of standard chemotherapy, median PFS was 4.7 months (95% CI: 4.5-5.1) and median OS was 11.2 months (95% CI: 9.9-13.8). Eight (35%) and three (13%) patients experienced at least one grade 3/4 or grade 4 AE, respectively. Commonly occurring grade 3/4 adverse events were thrombocytopenia (13%), fatigue (9%), nausea (9%), and vomiting (9%). CONCLUSIONS Saracatinib at a dose of 175 mg/day by mouth is well tolerated. However, the PFS rate observed at the pre-planned interim analysis did not meet the criteria for additional enrollment.
Drug Metabolism and Pharmacokinetics | 2016
Chalirmporn Atasilp; Pichai Chansriwong; Ekapob Sirachainan; Thanyanan Reungwetwattana; Montri Chamnanphon; Apichaya Puangpetch; Sansanee Wongwaisayawan; Chonlaphat Sukasem
UDP-glucuronosyltransferase1A1 (UGT1A1) polymorphisms have been related with irinotecan toxicity. The purpose of this study was to determine the associations between UGT1A1(*)28 and (*)6 polymorphisms and irinotecan toxicity in Thai patients with metastatic colorectal cancer. 44 metastatic colorectal cancer patients received irinotecan-based chemotherapy. Hematologic toxicities were determined in the first and second cycles of treatment. The genotypes of UGT1A1(*)28 and (*)6 were analyzed by pyrosequencing technique. The frequencies of genetic testing for UGT1A1(*)28 and (*)6 polymorphisms were 22.8% (TA6/TA7; 20.5%, TA7/TA7; 2.3%) and 15.9% (GA), respectively. No patients had the homozygous UGT1A1(*)6 (AA). Neither UGT1A1(*)28 nor UGT1A1(*)6 polymorphisms were significantly associated with severe hematologic toxicities. However, analysis of UGT1A1(*)28 and (*)6 in combination revealed an association with severe neutropenia in the first and second cycles (P = 0.044, P = 0.017, respectively). Both UGT1A1(*)28 and (*)6 polymorphisms may have an increased risk of irinotecan-induced neutropenia in Thai colorectal cancer patients.
Lung Cancer | 2017
Thanyanan Reungwetwattana; Ying Liang; Viola W. Zhu; Sai-Hong Ignatius Ou
A number of small molecule tyrosine kinase inhibitors (TKIs) have now been approved for the treatment of non-small cell lung cancers (NSCLC), including those targeted against epidermal growth factor receptor, anaplastic lymphoma kinase, and ROS1. Despite a wealth of agents developed to target the receptor tyrosine kinase, MET, clinical outcomes have as yet been disappointing, leading to pessimism about the role of MET in the pathogenesis of NSCLC. However, in recent years, there has been a renewed interest in MET exon 14 alterations as potential drivers of lung cancer. MET exon 14 alterations, which result in increased MET protein levels due to disrupted ubiquitin-mediated degradation, occur at a prevalence of around 3% in adenocarcinomas and around 2% in other lung neoplasms, making them attractive targets for the treatment of lung cancer. At least five MET-targeted TKIs, including crizotinib, cabozantinib, capmatinib, tepotinib, and glesatinib, are being investigated clinically for patients with MET exon 14 altered-NSCLC. A further two compounds have shown activity in preclinical models. In this article, we review the current clinical and preclinical data available for these TKIs, along with a number of other potential therapeutic options, including antibodies and immunotherapy. A number of questions remain unanswered regarding the future of MET TKIs, but unfortunately, the development of resistance to targeted therapies is inevitable. Resistance is expected to arise as a result of receptor tyrosine kinase mutation or from upregulation of MET ligand expression; potential strategies to overcome resistance are proposed.
Journal of Thoracic Oncology | 2018
Amanda McCambridge; Andrea Napolitano; Aaron S. Mansfield; Dean A. Fennell; Yoshitaka Sekido; Anna K. Nowak; Thanyanan Reungwetwattana; Weimin Mao; Harvey I. Pass; Michele Carbone; Haining Yang; Tobias Peikert
ABSTRACT Malignant pleural mesothelioma (MPM) is an uncommon, almost universally fatal, asbestos‐induced malignancy. New and effective strategies for diagnosis, prognostication, and treatment are urgently needed. Herein we review the advances in MPM achieved in 2017. Whereas recent epidemiological data demonstrated that the incidence of MPM‐related death continued to increase in United States between 2009 and 2015, new insight into the molecular pathogenesis and the immunological tumor microenvironment of MPM, for example, regarding the role of BRCA1 associated protein 1 and the expression programmed death receptor ligand 1, are highlighting new potential therapeutic strategies. Furthermore, there continues to be an ever‐expanding number of clinical studies investigating systemic therapies for MPM. These trials are primarily focused on immunotherapy using immune checkpoint inhibitors alone or in combination with other immunotherapies and nonimmunotherapies. In addition, other promising targeted therapies, including pegylated adenosine deiminase (ADI‐PEG20), which focuses on argininosuccinate synthase 1–deficient tumors, and tazemetostat, an enhancer of zeste 2 polycomb repressive complex 2 subunit inhibitor of BRCA1 associated protein 1 gene (BAP1)‐deficient tumors, are currently being explored.Malignant pleural mesothelioma (MPM) is an uncommon, almost universally fatal, asbestos-induced malignancy. New and effective strategies for diagnosis, prognostication and treatment are urgently needed. Herein we review the advances in MPM achieved in 2017. While recent epidemiological data demonstrated that the incidence of MPM-related death continued to increase in United States between 2009 and 2015, new insight into the molecular pathogenesis and the immunological tumor microenvironment of MPM, for example, regarding the role of BRCA1 associated protein 1 (BAP1) and the expression programmed death receptor ligand 1 (PD-L1), are highlighting new potential therapeutic strategies. Furthermore, there continues to be an ever-expanding number of clinical studies investigating systemic therapies for MPM. These trials are primarily focused on immunotherapy using immune checkpoint inhibitors alone or in combination with other immunoandnon-immuno therapies. In addition, other promising targeted therapies including ADI-PEG20 focusing on argininosuccinate synthase 1 deficient tumors and Tazemetostat, an EZH2 inhibitor of BAP1 deficient tumors are currently
Journal of Thoracic Oncology | 2016
Thanyanan Reungwetwattana; Alex A. Adjei
http://dx.doi.org/10.1016/j.jtho.2016.10.008 Cancer immunotherapy has led to a paradigm shift in the treatment of various types of malignancies in the last few years. Cancer cells are able to bypass host immunosurveillance by inhibiting antitumor lymphocytes and escaping tumor destruction. Immunosuppressive molecules such as cytotoxic T-lymphocyte associated antigen 4 (CTLA4), programmed cell death 1 (PD-1), and its ligand programmed death ligand 1 (PD-L1) are markedly overexpressed in the tumor microenvironment and lead to T-cell exhaustion. Targeting these molecules by immune checkpoint inhibitors, which are able to antagonize these immune-inhibitory pathways and restore cytotoxic T cells to destroy cancer cells, has shown significant clinical benefit. The first immune checkpoint inhibitor to be approved for clinical use was the anti-CTLA4 antibody ipilimumab. Since then, anti–PD-1 (nivolumab and pembrolizumab) and anti–PD-L1 (atezolizumab) agents have been approved for various types of cancers. By reactivating the immune system, these immune checkpoint inhibitors have led to the development of unusual immune-related adverse events (irAEs) in many organ systems, including the skin (rash/erythema and vitiligo), endocrine system (hypothyroidism/hyperthyroidism, thyroiditis, hypophysitis, diabetes mellitus, Addison’s disease, and adrenal insufficiency), gastrointestinal system (diarrhea, colitis, pancreatitis, and hepatitis), pulmonary system (pneumonitis and sarcoidosis), eyes and neurological system (episcleritis, uveitis, conjunctivitis, keratitis, orbital inflammation, myelitis, and aseptic meningitis), hematological system (red cell aplasia and autoimmune pancytopenia), renal system (interstitial nephritis, granulomatous nephritis, and glomerular lupus-like nephropathy), and musculoskeletal system (polyarthritis and arthralgia). These irAEs occur more frequently and are generally more severe in patients treated with an anti-CTLA4 agent than in those treated with anti–PD-1 and anti–PD-L1 agents. Rare irAEs such as Guillain-Barré syndrome, encephalopathy, aseptic meningitis, myasthenia gravis, myopathy, red cell aplasia, and acquired hemophilia A have been reported with ipilimumab treatment, whereas myasthenia gravis
Journal of Clinical Laboratory Analysis | 2018
Chalirmporn Atasilp; Pichai Chansriwong; Ekapob Sirachainan; Thanyanan Reungwetwattana; Apichaya Puangpetch; Santirhat Prommas; Suwannee Sirilerttrakul; Budsaba Rerkarmnuaychoke; Sansanee Wongwaisayawan; Chonlaphat Sukasem
Irinotecan (CPT‐11) is chemotherapy used mainly in the metastatic colorectal cancer. The purpose of this study was to develop and validate the LC‐MS/MS for the simultaneous determination of CPT‐11, SN‐38, and SN‐38G.
Journal of Pharmacogenomics and Pharmacoproteomics | 2012
Ekaphop Sirachainan; Thanyanan Reungwetwattana; Yupin Wisetpanit; Ravat Panvichian; Thitiya Sirisinha; Touch Ativitavas; Vorachai Ratanatharathorn; N. Trachu; Chonlaphat Sukasem
Objective: The objective of this study is to determine the extent of (DPYD) Dihydropyrimidine Dehydrogenase Gene] polymorphisms of Thai cancer patients who received 5-FU based chemotherapy regimens. Methods: The study was conducted a pharmacogenetic analysis to determine the polymorphisms of DPYD gene in 116. Thai cancer patients. 76 patients developed severe (grade 3-4) toxicities after receiving the first or second cycle of 5-FU based chemotherapy. The other subject group consisted of 40 patients without severe toxicity. The DNA sequencing of every amplicon was done to identify 11 mutations as reported in Asian population. The actual change of absolute neutrophil count (ANC), hematocrit, platelet and percentage of neutrophil were compared. Results: We detected 13 SNPs of which 6 SNPs were found in exons; 967G>A, 1011A>T, 1236G>A, 1774C>T, 1896T>C and 1627A>G. The other 7 SNPs were found in intron but only IVS14+1G>A is the intron splice site. We found homozygous GG of 1627A>G in 4 patients who had severe toxicities. Statistically significant difference in actual ANC change and percentage of neutrophil change in homozygous GG [P = .011 and .009] were found. The median nadir ANC of homozygous GG is 399.6 cells/mm3. This SNP has cause the amino acid change from isoleucine to valine. Novel heterozygous SNPs (967G>A, 1774C>T) that cause the amino acid change were found in two patients with severe toxicities. Conclusions: 1627A>G, 967G>A, 1774C>T and IVS14+G>A might be the cause of (DPD) Dihydro Pyrimidine Dehydrogenase deficiency in Thai patients. The further study needs to establish the functional DPD protein in this population. Ten novel SNPs were discovered in our study.