scispace - formally typeset
Search or ask a question
Author

Chung-Han Lee

Bio: Chung-Han Lee is an academic researcher from Memorial Sloan Kettering Cancer Center. The author has contributed to research in topics: Renal cell carcinoma & Medicine. The author has an hindex of 20, co-authored 68 publications receiving 2865 citations. Previous affiliations of Chung-Han Lee include Cornell University & University of Michigan.

Papers published on a yearly basis

Papers
More filters
Journal ArticleDOI
Robert M. Samstein1, Chung-Han Lee2, Chung-Han Lee1, Alexander N. Shoushtari1, Alexander N. Shoushtari2, Matthew D. Hellmann2, Matthew D. Hellmann1, Ronglai Shen1, Yelena Y. Janjigian1, Yelena Y. Janjigian2, David Barron1, Ahmet Zehir1, Emmet Jordan1, Antonio Omuro1, Thomas Kaley1, Sviatoslav M. Kendall1, Robert J. Motzer1, Robert J. Motzer2, A. Ari Hakimi1, Martin H. Voss2, Martin H. Voss1, Paul Russo1, Jonathan E. Rosenberg2, Jonathan E. Rosenberg1, Gopa Iyer1, Gopa Iyer2, Bernard H. Bochner1, Dean F. Bajorin1, Dean F. Bajorin2, Hikmat Al-Ahmadie1, Jamie E. Chaft1, Jamie E. Chaft2, Charles M. Rudin2, Charles M. Rudin1, Gregory J. Riely1, Gregory J. Riely2, Shrujal S. Baxi2, Shrujal S. Baxi1, Alan L. Ho1, Alan L. Ho2, Richard J. Wong1, David G. Pfister2, David G. Pfister1, Jedd D. Wolchok2, Jedd D. Wolchok1, Christopher A. Barker1, Philip H. Gutin1, Cameron Brennan1, Viviane Tabar1, Ingo K. Mellinghoff1, Lisa M. DeAngelis1, Charlotte E. Ariyan1, Nancy Y. Lee1, William D. Tap2, William D. Tap1, Mrinal M. Gounder2, Mrinal M. Gounder1, Sandra P. D'Angelo2, Sandra P. D'Angelo1, Leonard B. Saltz1, Leonard B. Saltz2, Zsofia K. Stadler2, Zsofia K. Stadler1, Howard I. Scher1, Howard I. Scher2, José Baselga2, José Baselga1, Pedram Razavi2, Pedram Razavi1, Christopher A. Klebanoff1, Christopher A. Klebanoff2, Rona Yaeger2, Rona Yaeger1, Neil H. Segal2, Neil H. Segal1, Geoffrey Y. Ku1, Geoffrey Y. Ku2, Ronald P. DeMatteo1, Marc Ladanyi1, Naiyer A. Rizvi3, Michael F. Berger1, Nadeem Riaz1, David B. Solit1, Timothy A. Chan1, Luc G. T. Morris1 
TL;DR: Analysis of advanced cancer patients treated with immune-checkpoint inhibitors shows that tumor mutational burden, as assessed by targeted next-generation sequencing, predicts survival after immunotherapy across multiple cancer types.
Abstract: Immune checkpoint inhibitor (ICI) treatments benefit some patients with metastatic cancers, but predictive biomarkers are needed. Findings in selected cancer types suggest that tumor mutational burden (TMB) may predict clinical response to ICI. To examine this association more broadly, we analyzed the clinical and genomic data of 1,662 advanced cancer patients treated with ICI, and 5,371 non-ICI-treated patients, whose tumors underwent targeted next-generation sequencing (MSK-IMPACT). Among all patients, higher somatic TMB (highest 20% in each histology) was associated with better overall survival. For most cancer histologies, an association between higher TMB and improved survival was observed. The TMB cutpoints associated with improved survival varied markedly between cancer types. These data indicate that TMB is associated with improved survival in patients receiving ICI across a wide variety of cancer types, but that there may not be one universal definition of high TMB.

2,343 citations

Journal ArticleDOI
TL;DR: An analytic pipeline and visualization tool (metabolograms) is developed to bridge the gap between TCGA transcriptomic profiling and metabolomic data, which enables to assemble an integrated pathway-level metabolic atlas and to demonstrate discordance between transcriptome and metabolome.

477 citations

Journal ArticleDOI
TL;DR: Lenvatinib plus pembrolizumab demonstrated a manageable safety profile and promising antitumor activity in patients with selected solid tumor types.
Abstract: PURPOSEModulation of vascular endothelial growth factor–mediated immune suppression via angiogenesis inhibition may augment the activity of immune checkpoint inhibitors. We report results from the ...

241 citations

Journal ArticleDOI
TL;DR: The mTOR pathway involves two functional complexes, TORC1 and TORC2, which have been defined by both their association with raptor or rictor, respectively, and their sensitivity to short-term rapamycin inhibition.
Abstract: Recent work has shown that the mTOR (mammalian target of rapamycin) pathway is an integral cell growth regulator. The mTOR pathway involves two functional complexes, TORC1 and TORC2, which have been defined by both their association with raptor or rictor, respectively, and their sensitivity to short-term rapamycin inhibition. Loss of tumor suppressors TSC1 or TSC2 leads to aberrant activation of TORC1, which has been implicated in the control of cell size. As a result, both physiologic and pathologic tissue hypertrophy are associated with TORC1 activation. Some clinical examples include skeletal and cardiac muscle hypertrophy, vascular restenosis, and compensatory nephrotic hypertrophy. Clarification of the mTOR pathway may lead to increased understanding of both the etiology and consequences of aberrant cell size regulation. This review covers some of the biochemical regulation of the mTOR pathway that may be important to the regulation of cell size, and it will present several potential clinical applications where the control of cell size may be biologically significant.

177 citations

Journal ArticleDOI
TL;DR: The coordinated relationship between mTOR and p53 during cellular stress provides a possible explanation for the benign nature of hamartoma syndromes, including TSC, and suggests that the efficacy of mTOR inhibitors in anti‐neoplastic therapy may also depend on p53 status, and m TOR inhibitors may antagonize the effects of genotoxic chemotherapeutics.
Abstract: Miscoordination of growth and proliferation with the cellular stress response can lead to tumorigenesis. Mammalian target of rapamycin (mTOR), a central cell growth controller, is highly activated in some malignant neoplasms, and its clinical implications are under extensive investigation. We show that constitutive mTOR activity amplifies p53 activation, in vitro and in vivo, by stimulating p53 translation. Thus, loss of TSC1 or TSC2, the negative regulators of mTOR, results in dramatic accumulation of p53 and apoptosis in response to stress conditions. In other words, the inactivation of mTOR prevents cell death by nutrient stress and genomic damage via p53. Consistently, we also show that p53 is elevated in TSC tumors, which rarely become malignant. The coordinated relationship between mTOR and p53 during cellular stress provides a possible explanation for the benign nature of hamartoma syndromes, including TSC. Clinically, this also suggests that the efficacy of mTOR inhibitors in anti-neoplastic therapy may also depend on p53 status, and mTOR inhibitors may antagonize the effects of genotoxic chemotherapeutics.

161 citations


Cited by
More filters
Journal ArticleDOI
TL;DR: AMPK directly phosphorylates the mTOR binding partner raptor on two well-conserved serine residues, and this phosphorylation induces 14-3-3 binding to raptor, uncovering a conserved effector of AMPK that mediates its role as a metabolic checkpoint coordinating cell growth with energy status.

3,328 citations

Journal ArticleDOI
TL;DR: The immunophenoscore was a superior predictor of response to anti-cytotoxic T lymphocyte antigen-4 (CTLA-4) and anti-programmed cell death protein 1 (anti-PD-1) antibodies in two independent validation cohorts and may help inform cancer immunotherapy and facilitate the development of precision immuno-oncology.

2,292 citations

Journal ArticleDOI
TL;DR: First-line treatment with nivolumab plus ipilimumab resulted in a longer duration of overall survival than did chemotherapy in patients with NSCLC, independent of the PD-L1 expression level.
Abstract: Background In an early-phase study involving patients with advanced non–small-cell lung cancer (NSCLC), the response rate was better with nivolumab plus ipilimumab than with nivolumab mono...

1,588 citations

Journal ArticleDOI
10 Aug 2017-Cell
TL;DR: A perspective on the roles of class I PI3Ks in the regulation of cellular metabolism and in immune system functions is provided, two topics closely intertwined with cancer biology.

1,461 citations

Journal ArticleDOI
TL;DR: A better understanding of how these variables cooperate to affect tumour–host interactions is needed to optimize the implementation of precision immunotherapy.
Abstract: Checkpoint inhibitor-based immunotherapies that target cytotoxic T lymphocyte antigen 4 (CTLA4) or the programmed cell death 1 (PD1) pathway have achieved impressive success in the treatment of different cancer types. Yet, only a subset of patients derive clinical benefit. It is thus critical to understand the determinants driving response, resistance and adverse effects. In this Review, we discuss recent work demonstrating that immune checkpoint inhibitor efficacy is affected by a combination of factors involving tumour genomics, host germline genetics, PD1 ligand 1 (PDL1) levels and other features of the tumour microenvironment, as well as the gut microbiome. We focus on recently identified molecular and cellular determinants of response. A better understanding of how these variables cooperate to affect tumour-host interactions is needed to optimize the implementation of precision immunotherapy.

1,452 citations