Try a new search

Format these results:

Searched for:

in-biosketch:true

person:laus09

Total Results:

28


Tarlatamab in Small-Cell Lung Cancer after Platinum-Based Chemotherapy

Mountzios, Giannis; Sun, Longhua; Cho, Byoung Chul; Demirci, Umut; Baka, Sofia; Gümüş, Mahmut; Lugini, Antonio; Zhu, Bo; Yu, Yan; Korantzis, Ippokratis; Han, Ji-Youn; Ciuleanu, Tudor-Eliade; Ahn, Myung-Ju; Rocha, Pedro; Mazières, Julien; Lau, Sally C M; Schuler, Martin; Blackhall, Fiona; Yoshida, Tatsuya; Owonikoko, Taofeek K; Paz-Ares, Luis; Jiang, Tony; Hamidi, Ali; Gauto, Diana; Recondo, Gonzalo; Rudin, Charles M; ,
BACKGROUND:Tarlatamab, a bispecific delta-like ligand 3-directed T-cell engager immunotherapy, received accelerated approval for the treatment of patients with previously treated small-cell lung cancer. Whether tarlatamab is more effective than chemotherapy in the treatment of patients whose small-cell lung cancer has progressed during or after initial platinum-based chemotherapy is not known. METHODS:We conducted a multinational, phase 3, open-label trial to compare tarlatamab with chemotherapy as second-line treatment in patients with small-cell lung cancer whose disease had progressed during or after platinum-based chemotherapy. Patients were randomly assigned to receive tarlatamab or chemotherapy (topotecan, lurbinectedin, or amrubicin). The primary end point was overall survival. Key secondary end points were investigator-assessed progression-free survival and patient-reported outcomes. Results of the prespecified interim analysis (data-cutoff date, January 29, 2025) are reported. RESULTS:A total of 509 patients were randomly assigned to receive tarlatamab (254 patients) or chemotherapy (255 patients). Treatment with tarlatamab resulted in significantly longer overall survival than chemotherapy (median, 13.6 months [95% confidence interval {CI}, 11.1 to not reached] vs. 8.3 months [95% CI, 7.0 to 10.2]; stratified hazard ratio for death, 0.60; 95% CI, 0.47 to 0.77; P<0.001). Tarlatamab treatment also had a significant benefit with respect to progression-free survival and cancer-related dyspnea and cough as compared with chemotherapy. The incidence of adverse events of grade 3 or higher was lower with tarlatamab than with chemotherapy (54% vs. 80%), as was the incidence of adverse events resulting in treatment discontinuation (5% vs. 12%). CONCLUSIONS:Treatment with tarlatamab led to longer overall survival than chemotherapy among patients with small-cell lung cancer whose disease had progressed during or after platinum-based chemotherapy. (Funded by Amgen; DeLLphi-304 ClinicalTrials.gov number, NCT05740566.).
PMID: 40454646
ISSN: 1533-4406
CID: 5862042

Author Correction: CTLA4 blockade abrogates KEAP1/STK11-related resistance to PD-(L)1 inhibitors

Skoulidis, Ferdinandos; Araujo, Haniel A; Do, Minh Truong; Qian, Yu; Sun, Xin; Cobo, Ana Galan; Le, John T; Montesion, Meagan; Palmer, Rachael; Jahchan, Nadine; Juan, Joseph M; Min, Chengyin; Yu, Yi; Pan, Xuewen; Arbour, Kathryn C; Vokes, Natalie; Schmidt, Stephanie T; Molkentine, David; Owen, Dwight H; Memmott, Regan; Patil, Pradnya D; Marmarelis, Melina E; Awad, Mark M; Murray, Joseph C; Hellyer, Jessica A; Gainor, Justin F; Dimou, Anastasios; Bestvina, Christine M; Shu, Catherine A; Riess, Jonathan W; Blakely, Collin M; Pecot, Chad V; Mezquita, Laura; Tabbó, Fabrizio; Scheffler, Matthias; Digumarthy, Subba; Mooradian, Meghan J; Sacher, Adrian G; Lau, Sally C M; Saltos, Andreas N; Rotow, Julia; Johnson, Rocio Perez; Liu, Corinne; Stewart, Tyler; Goldberg, Sarah B; Killam, Jonathan; Walther, Zenta; Schalper, Kurt; Davies, Kurtis D; Woodcock, Mark G; Anagnostou, Valsamo; Marrone, Kristen A; Forde, Patrick M; Ricciuti, Biagio; Venkatraman, Deepti; Van Allen, Eliezer M; Cummings, Amy L; Goldman, Jonathan W; Shaish, Hiram; Kier, Melanie; Katz, Sharyn; Aggarwal, Charu; Ni, Ying; Azok, Joseph T; Segal, Jeremy; Ritterhouse, Lauren; Neal, Joel W; Lacroix, Ludovic; Elamin, Yasir Y; Negrao, Marcelo V; Le, Xiuning; Lam, Vincent K; Lewis, Whitney E; Kemp, Haley N; Carter, Brett; Roth, Jack A; Swisher, Stephen; Lee, Richard; Zhou, Teng; Poteete, Alissa; Kong, Yifan; Takehara, Tomohiro; Paula, Alvaro Guimaraes; Parra Cuentas, Edwin R; Behrens, Carmen; Wistuba, Ignacio I; Zhang, Jianjun; Blumenschein, George R; Gay, Carl; Byers, Lauren A; Gibbons, Don L; Tsao, Anne; Lee, J Jack; Bivona, Trever G; Camidge, D Ross; Gray, Jhannelle E; Leighl, Natasha B; Levy, Benjamin; Brahmer, Julie R; Garassino, Marina C; Gandara, David R; Garon, Edward B; Rizvi, Naiyer A; Scagliotti, Giorgio Vittorio; Wolf, Jürgen; Planchard, David; Besse, Benjamin; Herbst, Roy S; Wakelee, Heather A; Pennell, Nathan A; Shaw, Alice T; Jänne, Pasi A; Carbone, David P; Hellmann, Matthew D; Rudin, Charles M; Albacker, Lee; Mann, Helen; Zhu, Zhou; Lai, Zhongwu; Stewart, Ross; Peters, Solange; Johnson, Melissa L; Wong, Kwok K; Huang, Alan; Winslow, Monte M; Rosen, Michael J; Winters, Ian P; Papadimitrakopoulou, Vassiliki A; Cascone, Tina; Jewsbury, Philip; Heymach, John V
PMID: 40016449
ISSN: 1476-4687
CID: 5801292

Practical management of adverse events in patients receiving tarlatamab, a delta-like ligand 3-targeted bispecific T-cell engager immunotherapy, for previously treated small cell lung cancer

Sands, Jacob M; Champiat, Stéphane; Hummel, Horst-Dieter; Paulson, Kelly G; Borghaei, Hossein; Alvarez, Jean Bustamante; Carbone, David P; Carlisle, Jennifer W; Choudhury, Noura J; Clarke, Jeffrey M; Gadgeel, Shirish M; Izumi, Hiroki; Navarro, Alejandro; Lau, Sally C M; Lammers, Philip E; Huang, Shuang; Hamidi, Ali; Mukherjee, Sujoy; Owonikoko, Taofeek K
Tarlatamab is a bispecific T-cell engager immunotherapy targeting delta-like ligand 3 (DLL3) and the cluster of differentiation 3 (CD3) molecule. In the phase 2 DeLLphi-301 trial of tarlatamab for patients with previously treated small cell lung cancer, tarlatamab 10 mg every 2 weeks achieved durable responses and encouraging survival outcomes. Analyses of updated safety data from the DeLLphi-301 trial demonstrated that the most common treatment-emergent adverse events were cytokine release syndrome (53%), pyrexia (38%), decreased appetite (36%), dysgeusia (32%), and an emia (30%). Cytokine release syndrome was mostly grade 1 or 2 in severity, occurred primarily after the first or second tarlatamab dose, and was managed with supportive care, which included the administration of antipyretics (e.g., acetaminophen), intravenous hydration, and/or glucocorticoids. Other treatment-emergent adverse effects of interest included neutropenia (16%) and immune effector cell-associated neurotoxicity syndrome and associated neurologic events (10%). Given that tarlatamab is the first T-cell engager approved for the treatment of small cell lung cancer, raising awareness with regard to the monitoring and management of tarlatamab-associated adverse events is essential. Here, the authors describe the timing, occurrence, and duration of these adverse events and review the management and risk-mitigation strategies used by clinical investigators during the DeLLphi-301 trial.
PMCID:11775405
PMID: 39876075
ISSN: 1097-0142
CID: 5780842

CTLA4 blockade abrogates KEAP1/STK11-related resistance to PD-(L)1 inhibitors

Skoulidis, Ferdinandos; Araujo, Haniel A; Do, Minh Truong; Qian, Yu; Sun, Xin; Cobo, Ana Galan; Le, John T; Montesion, Meagan; Palmer, Rachael; Jahchan, Nadine; Juan, Joseph M; Min, Chengyin; Yu, Yi; Pan, Xuewen; Arbour, Kathryn C; Vokes, Natalie; Schmidt, Stephanie T; Molkentine, David; Owen, Dwight H; Memmott, Regan; Patil, Pradnya D; Marmarelis, Melina E; Awad, Mark M; Murray, Joseph C; Hellyer, Jessica A; Gainor, Justin F; Dimou, Anastasios; Bestvina, Christine M; Shu, Catherine A; Riess, Jonathan W; Blakely, Collin M; Pecot, Chad V; Mezquita, Laura; Tabbó, Fabrizio; Scheffler, Matthias; Digumarthy, Subba; Mooradian, Meghan J; Sacher, Adrian G; Lau, Sally C M; Saltos, Andreas N; Rotow, Julia; Johnson, Rocio Perez; Liu, Corinne; Stewart, Tyler; Goldberg, Sarah B; Killam, Jonathan; Walther, Zenta; Schalper, Kurt; Davies, Kurtis D; Woodcock, Mark G; Anagnostou, Valsamo; Marrone, Kristen A; Forde, Patrick M; Ricciuti, Biagio; Venkatraman, Deepti; Van Allen, Eliezer M; Cummings, Amy L; Goldman, Jonathan W; Shaish, Hiram; Kier, Melanie; Katz, Sharyn; Aggarwal, Charu; Ni, Ying; Azok, Joseph T; Segal, Jeremy; Ritterhouse, Lauren; Neal, Joel W; Lacroix, Ludovic; Elamin, Yasir Y; Negrao, Marcelo V; Le, Xiuning; Lam, Vincent K; Lewis, Whitney E; Kemp, Haley N; Carter, Brett; Roth, Jack A; Swisher, Stephen; Lee, Richard; Zhou, Teng; Poteete, Alissa; Kong, Yifan; Takehara, Tomohiro; Paula, Alvaro Guimaraes; Parra Cuentas, Edwin R; Behrens, Carmen; Wistuba, Ignacio I; Zhang, Jianjun; Blumenschein, George R; Gay, Carl; Byers, Lauren A; Gibbons, Don L; Tsao, Anne; Lee, J Jack; Bivona, Trever G; Camidge, D Ross; Gray, Jhannelle E; Lieghl, Natasha; Levy, Benjamin; Brahmer, Julie R; Garassino, Marina C; Gandara, David R; Garon, Edward B; Rizvi, Naiyer A; Scagliotti, Giorgio Vittorio; Wolf, Jürgen; Planchard, David; Besse, Benjamin; Herbst, Roy S; Wakelee, Heather A; Pennell, Nathan A; Shaw, Alice T; Jänne, Pasi A; Carbone, David P; Hellmann, Matthew D; Rudin, Charles M; Albacker, Lee; Mann, Helen; Zhu, Zhou; Lai, Zhongwu; Stewart, Ross; Peters, Solange; Johnson, Melissa L; Wong, Kwok K; Huang, Alan; Winslow, Monte M; Rosen, Michael J; Winters, Ian P; Papadimitrakopoulou, Vassiliki A; Cascone, Tina; Jewsbury, Philip; Heymach, John V
For patients with advanced non-small-cell lung cancer (NSCLC), dual immune checkpoint blockade (ICB) with CTLA4 inhibitors and PD-1 or PD-L1 inhibitors (hereafter, PD-(L)1 inhibitors) is associated with higher rates of anti-tumour activity and immune-related toxicities, when compared with treatment with PD-(L)1 inhibitors alone. However, there are currently no validated biomarkers to identify which patients will benefit from dual ICB1,2. Here we show that patients with NSCLC who have mutations in the STK11 and/or KEAP1 tumour suppressor genes derived clinical benefit from dual ICB with the PD-L1 inhibitor durvalumab and the CTLA4 inhibitor tremelimumab, but not from durvalumab alone, when added to chemotherapy in the randomized phase III POSEIDON trial3. Unbiased genetic screens identified loss of both of these tumour suppressor genes as independent drivers of resistance to PD-(L)1 inhibition, and showed that loss of Keap1 was the strongest genomic predictor of dual ICB efficacy-a finding that was confirmed in several mouse models of Kras-driven NSCLC. In both mouse models and patients, KEAP1 and STK11 alterations were associated with an adverse tumour microenvironment, which was characterized by a preponderance of suppressive myeloid cells and the depletion of CD8+ cytotoxic T cells, but relative sparing of CD4+ effector subsets. Dual ICB potently engaged CD4+ effector cells and reprogrammed the tumour myeloid cell compartment towards inducible nitric oxide synthase (iNOS)-expressing tumoricidal phenotypes that-together with CD4+ and CD8+ T cells-contributed to anti-tumour efficacy. These data support the use of chemo-immunotherapy with dual ICB to mitigate resistance to PD-(L)1 inhibition in patients with NSCLC who have STK11 and/or KEAP1 alterations.
PMID: 39385035
ISSN: 1476-4687
CID: 5730202

Top 20 EGFR+ NSCLC Clinical and Translational Science Papers That Shaped the 20 Years Since the Discovery of Activating EGFR Mutations in NSCLC. An Editor-in-Chief Expert Panel Consensus Survey

Ou, Sai-Hong Ignatius; Le, Xiuning; Nagasaka, Misako; Reungwetwattana, Thanyanan; Ahn, Myung-Ju; Lim, Darren W T; Santos, Edgardo S; Shum, Elaine; Lau, Sally C M; Lee, Jii Bum; Calles, Antonio; Wu, Fengying; Lopes, Gilberto; Sriuranpong, Virote; Tanizaki, Junko; Horinouchi, Hidehito; Garassino, Marina C; Popat, Sanjay; Besse, Benjamin; Rosell, Rafael; Soo, Ross A
The year 2024 is the 20th anniversary of the discovery of activating epidermal growth factor receptor (EGFR) mutations in non-small cell lung cancer (NSCLC). Since then, tremendous advances have been made in the treatment of NSCLC based on this discovery. Some of these studies have led to seismic changes in the concept of oncology research and spurred treatment advances beyond NSCLC, leading to a current true era of precision oncology for all solid tumors. We now routinely molecularly profile all tumor types and even plasma samples of patients with NSCLC for multiple actionable driver mutations, independent of patient clinical characteristics nor is profiling limited to the advanced incurable stage. We are increasingly monitoring treatment responses and detecting resistance to targeted therapy by using plasma genotyping. Furthermore, we are now profiling early-stage NSCLC for appropriate adjuvant targeted treatment leading to an eventual potential "cure" in early-stage EGFR+ NSCLC which have societal implication on implementing lung cancer screening in never-smokers as most EGFR+ NSCLC patients are never-smokers. All these advances were unfathomable in 2004 when the five papers that described "discoveries" of activating EGFR mutations (del19, L858R, exon 20 insertions, and "uncommon" mutations) were published. To commemorate this 20th anniversary, we assembled a global panel of thoracic medical oncology experts to select the top 20 papers (publications or congress presentation) from the 20 years since this seminal discovery with December 31, 2023 as the cutoff date for inclusion of papers to be voted on. Papers ranked 21 to 30 were considered "honorable mention" and also annotated. Our objective is that these 30 papers with their annotations about their impact and even all the ranked papers will serve as "syllabus" for the education of future thoracic oncology trainees. Finally, we mentioned potential practice-changing clinical trials to be reported. One of them, LAURA was published online on June 2, 2024 was not included in the list of papers to be voted on but will surely be highly ranked if this consensus survery is performed again on the 25th anniversay of the discovery EGFR mutations (i.e. top 25 papers on the 25 years since the discovery of activating EGFR mutations).
PMCID:11208875
PMID: 38938224
ISSN: 1179-2728
CID: 5733402

Programmed Cell Death Protein 1 Inhibitors and MET Targeted Therapies in NSCLC With MET Exon 14 Skipping Mutations: Efficacy and Toxicity as Sequential Therapies

Lau, Sally C M; Perdrizet, Kirstin; Fung, Andrea S; Mata, Danilo Giffoni M M; Weiss, Jessica; Holzapfel, Nick; Liu, Geoffrey; Bradbury, Penelope A; Shepherd, Frances A; Sacher, Adrian G; Feilotter, Harriet; Sheffield, Brandon; Hwang, David; Tsao, Ming Sound; Cheng, Susanna; Cheema, Parneet; Leighl, Natasha B
INTRODUCTION/UNASSIGNED:ex14-mutant NSCLC. METHODS/UNASSIGNED:ex14 alterations between 2014 and 2020 across four Canadian cancer centers. Demographics, disease characteristics, systemic therapy, overall response rates (ORRs), survival, and toxicity were summarized. RESULTS/UNASSIGNED:ex14-mutant NSCLC, the median overall survival was 23.1 months: 127.0 months in stage 1, 27.3 months in resected stage 2 and 3, and 16.6 months in unresectable stage 3 or 4 disease, respectively. In patients with advanced disease, 22% were too unwell for systemic treatment. MET tyrosine kinase inhibitors (TKIs) were administered to 28 patients with an ORR of 33%, median progression-free survival of 2.7 months, and 3.8 months for selective TKIs. Programmed cell death protein-1 (PD-1) inhibitors were given to 25 patients-the ORR was 44% and progression-free survival was 10.6 months. No responses were seen with subsequent MET TKIs after initial TKI treatment. Grade 3 or higher toxicities occurred in 64% of patients who received MET TKI after PD-1 inhibitors versus 8% in those who did not receive PD-1 inhibitors. CONCLUSIONS/UNASSIGNED:ex14 skipping and initial therapy are imperative to improving patient survival.
PMCID:10514105
PMID: 37744308
ISSN: 2666-3643
CID: 5725172

Spotlight on Small-Cell Lung Cancer and Other Lung Neuroendocrine Neoplasms

Fernandez-Cuesta, Lynnette; Sexton-Oates, Alexandra; Bayat, Leyla; Foll, Matthieu; Lau, Sally C M; Leal, Ticiana
Lung neuroendocrine neoplasms (NENs) encompass a spectrum of neoplasms that are subdivided into the well-differentiated neuroendocrine tumors comprising the low- and intermediate-grade typical and atypical carcinoids, respectively, and the poorly differentiated, high-grade neuroendocrine carcinomas including large-cell neuroendocrine carcinomas and small-cell lung carcinoma (SCLC). Here, we review the current morphological and molecular classifications of the NENs on the basis of the updated WHO Classification of Thoracic Tumors and discuss the emerging subclassifications on the basis of molecular profiling and the potential therapeutic implications. We focus on the efforts in subtyping SCLC, a particularly aggressive tumor with few treatment options, and the recent advances in therapy with the adoption of immune checkpoint inhibitors in the frontline setting for patients with extensive-stage SCLC. We further highlight the promising immunotherapy strategies in SCLC that are currently under investigation.
PMID: 37229617
ISSN: 1548-8756
CID: 5508552

Selpercatinib as the Guardian of the Central Nervous System for Patients With RET Fusion-Positive NSCLC? [Editorial]

Lau, Sally C M; Ou, Sai-Hong Ignatius
PMID: 37087118
ISSN: 1556-1380
CID: 5464672

In vivo metabolomics identifies CD38 as an emergent vulnerability in LKB1 -mutant lung cancer

Deng, Jiehui; Peng, David H; Fenyo, David; Yuan, Hao; Lopez, Alfonso; Levin, Daniel S; Meynardie, Mary; Quinteros, Mari; Ranieri, Michela; Sahu, Soumyadip; Lau, Sally C M; Shum, Elaine; Velcheti, Vamsidhar; Punekar, Salman R; Rekhtman, Natasha; Dowling, Catríona M; Weerasekara, Vajira; Xue, Yun; Ji, Hongbin; Siu, Yik; Jones, Drew; Hata, Aaron N; Shimamura, Takeshi; Poirier, John T; Rudin, Charles M; Hattori, Takamitsu; Koide, Shohei; Papagiannakopoulos, Thales; Neel, Benjamin G; Bardeesy, Nabeel; Wong, Kwok-Kin
UNLABELLED:. Surprisingly, compared with other genetic subsets, murine and human LKB1-mutant NSCLC show marked overexpression of the NAD+-catabolizing ectoenzyme, CD38 on the surface of tumor cells. Loss of LKB1 or inactivation of Salt-Inducible Kinases (SIKs)-key downstream effectors of LKB1- induces CD38 transcription induction via a CREB binding site in the CD38 promoter. Treatment with the FDA-approved anti-CD38 antibody, daratumumab, inhibited growth of LKB1-mutant NSCLC xenografts. Together, these results reveal CD38 as a promising therapeutic target in patients with LKB1 mutant lung cancer. SIGNIFICANCE/CONCLUSIONS:tumor suppressor of lung adenocarcinoma patients and are associated with resistance to current treatments. Our study identified CD38 as a potential therapeutic target that is highly overexpressed in this specific subtype of cancer, associated with a shift in NAD homeostasis.
PMCID:10153147
PMID: 37131623
ISSN: 2692-8205
CID: 5507602

The Value of Comprehensive Molecular Profiling in Early Stage Lung Cancer [Editorial]

Lau, Sally C M
PMID: 36990570
ISSN: 1556-1380
CID: 5463332