429 results on '"Suresh S Ramalingam"'
Search Results
2. Five-Year Survival Outcomes With Nivolumab Plus Ipilimumab Versus Chemotherapy as First-Line Treatment for Metastatic Non–Small-Cell Lung Cancer in CheckMate 227
- Author
-
Julie R. Brahmer, Jong-Seok Lee, Tudor-Eliade Ciuleanu, Reyes Bernabe Caro, Makoto Nishio, Laszlo Urban, Clarisse Audigier-Valette, Lorena Lupinacci, Randeep Sangha, Adam Pluzanski, Jacobus Burgers, Mauricio Mahave, Samreen Ahmed, Adam J. Schoenfeld, Luis G. Paz-Ares, Martin Reck, Hossein Borghaei, Kenneth J. O'Byrne, Ravi G. Gupta, Judith Bushong, Li Li, Steven I. Blum, Laura J. Eccles, and Suresh S. Ramalingam
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE We present 5-year results from CheckMate 227 Part 1, in which nivolumab plus ipilimumab improved overall survival (OS) versus chemotherapy in patients with metastatic non–small-cell lung cancer, regardless of tumor programmed death ligand 1 (PD-L1) status. METHODS Adults with stage IV/recurrent non–small-cell lung cancer without EGFR mutations or ALK alterations and with tumor PD-L1 ≥ 1% or < 1% (n = 1739) were randomly assigned. Patients with tumor PD-L1 ≥ 1% were randomly assigned to first-line nivolumab plus ipilimumab, nivolumab alone, or chemotherapy. Patients with tumor PD-L1 < 1% were randomly assigned to nivolumab plus ipilimumab, nivolumab plus chemotherapy, or chemotherapy. End points included exploratory 5-year results for efficacy, safety, and quality of life. RESULTS At a minimum follow-up of 61.3 months, 5-year OS rates were 24% versus 14% for nivolumab plus ipilimumab versus chemotherapy (PD-L1 ≥ 1%) and 19% versus 7% (PD-L1 < 1%). The median duration of response was 24.5 versus 6.7 months (PD-L1 ≥ 1%) and 19.4 versus 4.8 months (PD-L1 < 1%). Among patients surviving 5 years, 66% (PD-L1 ≥ 1%) and 64% (PD-L1 < 1%) were off nivolumab plus ipilimumab without initiating subsequent systemic anticancer treatment by the 5-year time point. Survival benefit continued after nivolumab plus ipilimumab discontinuation because of treatment-related adverse events, with a 5-year OS rate of 39% (combined PD-L1 ≥ 1% and < 1% populations). Quality of life in 5-year survivors treated with nivolumab plus ipilimumab was similar to that in the general US population through the 5-year follow-up. No new safety signals were observed. CONCLUSION With all patients off immunotherapy treatment for ≥ 3 years, nivolumab plus ipilimumab increased 5-year survivorship versus chemotherapy, including long-term, durable clinical benefit regardless of tumor PD-L1 expression. These data support nivolumab plus ipilimumab as an effective first-line treatment for patients with metastatic non–small-cell lung cancer. [Media: see text]
- Published
- 2023
3. Engagement and outcomes of cancer patients referred to a tobacco cessation program at a National Cancer Institute‐designated cancer center
- Author
-
Sarah A. Westergaard, Manali Rupji, Lauren E. Franklin, Madhusmita Behera, Suresh S. Ramalingam, and Kristin A. Higgins
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Abstract
Tobacco cessation is a critical but challenging intervention for cancer patients. Our National Cancer Institute-designated Comprehensive Cancer Center instituted a tobacco cessation program in 2019. This manuscript reports on the first 2 years of our experience.Patients were referred to the program by their care team, and a certified tobacco treatment specialist contacted patients remotely and provided behavioral therapy and coordinated pharmacotherapy. We retrospectively captured data from patients with a cancer diagnosis referred to the tobacco cessation program. Univariate and multivariable logistic regression analyses with the backward elimination approach were performed to determine factors associated with patient acceptance of referral to the tobacco cessation program. Tobacco cessation rates after referral to the program were also captured.Between July 2019 and August 2021, 194 patients were referred to the tobacco cessation program. Of the 194 patients referred, 93 agreed to enroll in the tobacco cessation program (47.9%), of which 84 requested pharmacotherapy (90.3%). Twenty-four were able to cease tobacco use (25.8%). Only 7 patients out of the 101 patients (6.9%) who declined cessation services were successful (p 0.001). On univariate logistic regression, race (p = 0.027) and marital status (p = 0.020) were associated with referral acceptance. On multivariable analysis, single patients (odds ratio [OR] = 0.33) and Caucasian patients (OR = 0.43) were less likely to accept a referral.Access to tobacco cessation services is a critical component of comprehensive cancer care. Our experience highlights the need to understand patient-specific factors associated with engagement with a tobacco cessation program during cancer treatment. The use of pharmacotherapy is also a critical component of successful tobacco cessation.
- Published
- 2022
4. Long-Term Outcomes and Molecular Correlates of Sotorasib Efficacy in Patients With Pretreated KRAS G12C-Mutated Non-Small-Cell Lung Cancer: 2-Year Analysis of CodeBreaK 100
- Author
-
Grace K. Dy, Ramaswamy Govindan, Vamsidhar Velcheti, Gerald S. Falchook, Antoine Italiano, Jürgen Wolf, Adrian G. Sacher, Toshiaki Takahashi, Suresh S. Ramalingam, Christophe Dooms, Dong-Wan Kim, Alfredo Addeo, Jayesh Desai, Martin Schuler, Pascale Tomasini, David S. Hong, Piro Lito, Qui Tran, Simon Jones, Abraham Anderson, Antreas Hindoyan, Wendy Snyder, Ferdinandos Skoulidis, and Bob T. Li
- Subjects
Cancer Research ,Oncology - Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported.In the longest follow-up, to our knowledge, for a KRASG12C inhibitor, we assessed the long-term efficacy, safety, and biomarkers of sotorasib in patients with KRAS G12C-mutated advanced non-small-cell lung cancer (NSCLC) from the CodeBreaK 100 clinical trial (ClinicalTrials.gov identifier: NCT03600883). This multicenter, single-group, open-label phase I/phase II trial enrolled 174 patients with KRAS G12C-mutated, locally advanced or metastatic NSCLC after progression on prior therapies. Patients (N = 174) received sotorasib 960 mg once daily with the primary end points for phase I of safety and tolerability and for phase II of objective response rate (ORR). Sotorasib produced an ORR of 41%, median duration of response of 12.3 months, progression-free survival (PFS) of 6.3 months, overall survival (OS) of 12.5 months, and 2-year OS rate of 33%. Long-term clinical benefit (PFS ≥ 12 months) was observed in 40 (23%) patients across PD-L1 expression levels, in a proportion of patients with somatic STK11 and/or KEAP1 alterations, and was associated with lower baseline circulating tumor DNA levels. Sotorasib was well tolerated, with few late-onset treatment-related toxicities, none of which led to treatment discontinuation. These results demonstrate the long-term benefit of sotorasib, including in subgroups with poor prognosis. ispartof: J Clin Oncol vol:41 issue:18 pages:3311-3317 ispartof: location:United States status: published
- Published
- 2023
5. Cancer Turns 75!
- Author
-
Suresh S. Ramalingam
- Subjects
Cancer Research ,Oncology - Published
- 2023
6. Impact of concomitant fibrates on immunotherapy outcomes for advanced <scp>non‐small</scp> cell lung cancer
- Author
-
William A. Stokes, Madhusmita Behera, Renjian Jiang, David A. Gutman, Zhonglu Huang, Abigail Burns, Nikhil T. Sebastian, Vidula Sukhatme, Michael C. Lowe, Suresh S. Ramalingam, Vikas P. Sukhatme, and Drew Moghanaki
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Abstract
Peroxisome proliferator-activated receptor agonists such as fibrates restore oxidative metabolism in cytotoxic T-lymphocytes, thereby enhancing response to immune checkpoint inhibitors (ICI) in preclinical models. However, there is no evidence in humans on the clinical impact of fibrates as an adjunct to ICI.In this cohort study of Veterans with non-small cell lung cancer (NSCLC) receiving ICI, fibrate exposure was defined as a prescription filled within 90 days of an ICI infusion. Overall survival (OS), measured from the start of ICI, was compared between exposed and unexposed Veterans. Cox multivariable analysis (MVA) was used to identify factors associated with OS. A sensitivity analysis of Veterans with stage IV NSCLC who received docetaxel without ICI was similarly performed.The ICI cohort included 3593 Veterans, of whom 301 (8.5%) coincidentally received a fibrate. Veterans receiving fibrates were more likely to be older, white, male, and married, and to have greater comorbidity burden, but less likely to receive chemotherapy. Coincidental fibrates were associated with improved OS both on MVA (HR 0.86, 95%CI 0.75-0.99) and in a matched subset (HR 0.75, 95%CI 0.63-0.90). In contrast, among the cohort of 968 Veterans treated with chemotherapy, fibrates did not have a significant impact on OS by MVA (HR 0.99, 95%CI 0.79-1.25) or in a matched subset (HR 1.02, 95%CI CI 0.75-1.39).Concomitant fibrates are associated with improved OS among NSCLC patients receiving ICI but not among those receiving chemotherapy. This hypothesis-generating observation supports a potential role for fibrates as an adjunct to immunotherapy.
- Published
- 2022
7. Therapeutic efficacy of the novel SHP2 degrader SHP2-D26, alone or in combination, against lung cancer is associated with modulation of p70S6K/S6, Bim and Mcl-1
- Author
-
Yunfu Deng, Guangzhi Ma, Karin A. Vallega, Dongsheng Wang, Mingliang Wang, Changwei Wang, Shaomeng Wang, Suresh S. Ramalingam, and Shi-Yong Sun
- Subjects
ErbB Receptors ,Cancer Research ,Lung Neoplasms ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Mutation ,Humans ,Ribosomal Protein S6 Kinases, 70-kDa ,Molecular Medicine ,Protein Kinase Inhibitors ,Molecular Biology - Abstract
SHP2, a protein tyrosine phosphatase, plays a critical role in fully activating oncogenic signaling pathways such as Ras/MAPK downstream of cell surface tyrosine receptors (e.g., EGFR), which are often activated in human cancers, and thus has emerged as an attractive cancer therapeutic target. This study focused on evaluating the therapeutic potential of the novel SHP2 degrader, SHP2-D26 (D26), either alone or in combination, against non-small cell lung cancer (NSCLC) cells. While all tested NSCLC cell lines responded to D26 with IC
- Published
- 2022
8. Lung-MAP: A Collaborative Roadmap to Improve Cancer Outcomes
- Author
-
Ticiana A. Leal, Suzanne E. Dahlberg, and Suresh S. Ramalingam
- Subjects
Cancer Research ,Oncology - Published
- 2022
9. Spatially variant immune infiltration scoring in human cancer tissues
- Author
-
Mayar Allam, Thomas Hu, Jeongjin Lee, Jeffrey Aldrich, Sunil S. Badve, Yesim Gökmen-Polar, Manali Bhave, Suresh S. Ramalingam, Frank Schneider, and Ahmet F. Coskun
- Subjects
Cancer Research ,Oncology - Abstract
The Immunoscore is a method to quantify the immune cell infiltration within cancers to predict the disease prognosis. Previous immune profiling approaches relied on limited immune markers to establish patients’ tumor immunity. However, immune cells exhibit a higher-level complexity that is typically not obtained by the conventional immunohistochemistry methods. Herein, we present a spatially variant immune infiltration score, termed as SpatialVizScore, to quantify immune cells infiltration within lung tumor samples using multiplex protein imaging data. Imaging mass cytometry (IMC) was used to target 26 markers in tumors to identify stromal, immune, and cancer cell states within 26 human tissues from lung cancer patients. Unsupervised clustering methods dissected the spatial infiltration of cells in tissue using the high-dimensional analysis of 16 immune markers and other cancer and stroma enriched labels to profile alterations in the tumors’ immune infiltration patterns. Spatially resolved maps of distinct tumors determined the spatial proximity and neighborhoods of immune-cancer cell pairs. These SpatialVizScore maps provided a ranking of patients’ tumors consisting of immune inflamed, immune suppressed, and immune cold states, demonstrating the tumor’s immune continuum assigned to three distinct infiltration score ranges. Several inflammatory and suppressive immune markers were used to establish the cell-based scoring schemes at the single-cell and pixel-level, depicting the cellular spectra in diverse lung tissues. Thus, SpatialVizScore is an emerging quantitative method to deeply study tumor immunology in cancer tissues.
- Published
- 2022
10. Discovery of Small Molecule Bak Activator for Lung Cancer Therapy
- Author
-
Maohua Xie, Xingming Deng, Abu Syed Md Anisuzzaman, Andrew T. Magis, Suresh S. Ramalingam, Gabriel Sica, Guojing Zhang, Guo Chen, Taofeek K. Owonikoko, Dongkyoo Park, and Madhusmita Behera
- Subjects
Lung Neoplasms ,Medicine (miscellaneous) ,Mice, Nude ,Antineoplastic Agents ,Small Molecule Libraries ,chemistry.chemical_compound ,Mice ,Protein Domains ,Radioresistance ,Carcinoma, Non-Small-Cell Lung ,Proto-Oncogene Proteins ,medicine ,small molecule activator ,Animals ,Humans ,Cytotoxicity ,Lung cancer ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Death domain ,BH3 domain ,therapy ,Venetoclax ,Activator (genetics) ,Bak ,apoptosis ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Peptide Fragments ,Mitochondria ,lung cancer ,bcl-2 Homologous Antagonist-Killer Protein ,chemistry ,Proto-Oncogene Proteins c-bcl-2 ,Apoptosis ,Cancer research ,biological phenomena, cell phenomena, and immunity ,Research Paper ,Protein Binding - Abstract
Rationale: Bak is a major proapoptotic Bcl2 family member and a required molecule for apoptotic cell death. High levels of endogenous Bak were observed in both small cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC) cell lines. Increased Bak expression was correlated with poor prognosis of NSCLC patients, suggesting that Bak protein is an attractive target for lung cancer therapy. The BH3 domain functions as death domain and is required for Bak to initiate apoptotic cell death. Thus, the BH3 domain is attractive target for discovery of Bak agonist. Methods: The BH3 death domain binding pocket (aa75-88) of Bak was chosen as a docking site for screening of small molecule Bak activators using the UCSF DOCK 6.1 program suite and the NCI chemical library (300,000 small molecules) database. The top 500 compounds determined to have the highest affinity for the BH3 domain were obtained from the NCI and tested for cytotoxicity for further screening. We identified a small molecule Bak activator BKA-073 as the lead compound. The binding affinity of BKA-073 with Bak protein was analyzed by isothermal titration calorimetry (ITC) assay. BKA-073-mediated Bak activation via oligomerization was analyzed by a cross-linking with Bis (maleimido) hexane (BMH). Sensitivity of BKA-073 to lung cancer cells in vitro was evaluated by dynamic BH3 profiling (DBP) and apoptotic cell death assay. The potency of BKA-073 alone or in combination with radiotherapy or Bcl2 inhibitor was evaluated in animal models. Results: We found that BKA-073 binds Bak at BH3 domain with high affinity and selectivity. BKA-073/Bak binding promotes Bak oligomerization and mitochondrial priming that activates its proapoptotic function. BKA-073 potently suppresses tumor growth without significant normal tissue toxicity in small cell lung cancer (SCLC) and NSCLC xenografts, patient-derived xenografts, and genetically engineered mouse models of mutant KRAS-driven cancer. Bak accumulates in radioresistant lung cancer cells and BKA-073 reverses radioresistance. Combination of BKA-073 with Bcl-2 inhibitor venetoclax exhibits strong synergy against lung cancer in vivo. Conclusions: Development of small molecule Bak activator may provide a new class of anticancer agents to treat lung cancer.
- Published
- 2021
11. Membrane-Associated RING-CH 8 Functions as a Novel PD-L1 E3 Ligase to Mediate PD-L1 Degradation Induced by EGFR Inhibitors
- Author
-
Wenyi Wei, Songqing Fan, Guoqing Qian, Karin A. Vallega, Qiming Wang, Yunfu Deng, Zhen Chen, Suresh S. Ramalingam, Jianping Guo, Taofeek K. Owonikoko, Changjiang Hu, and Shi-Yong Sun
- Subjects
Cancer Research ,Lung Neoplasms ,Ubiquitin-Protein Ligases ,Mice, Nude ,Apoptosis ,B7-H1 Antigen ,Article ,Cell Line ,Glycogen Synthase Kinase 3 ,Mice ,Ubiquitin ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,PD-L1 ,Animals ,Humans ,Osimertinib ,Epidermal growth factor receptor ,Protein Kinase Inhibitors ,Molecular Biology ,EGFR inhibitors ,Acrylamides ,Gene knockdown ,Aniline Compounds ,biology ,Chemistry ,Ubiquitin ligase ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,HEK293 Cells ,Oncology ,Mutation ,Cancer cell ,biology.protein ,Cancer research ,Signal Transduction - Abstract
Expression of programmed death-ligand 1 (PD-L1) on cancer cells is a critical mechanism contributing to immunosuppression and immune escape. PD-L1 expression may also affect therapeutic outcomes of epidermal growth factor receptor (EGFR)-targeted therapy (e.g., with osimertinib/AZD9291) against EGFR-mutant non–small cell lung cancers (NSCLC) and can even be altered during the treatment albeit with largely undefined mechanisms. This study primarily focuses on elucidating the mechanism by which osimertinib induces PD-L1 degradation in addition to validating osimertinib's effect on decreasing PD-L1 expression in EGFR-mutant NSCLC cells and tumors. Osimertinib and other EGFR inhibitors effectively decreased PD-L1 levels primarily in EGFR-mutant NSCLCs and xenografted tumors. Osimertinib not only decreased PD-L1 mRNA expression, but also prompted proteasomal degradation of PD-L1 protein, indicating both transcriptional and posttranslational mechanisms accounting for osimertinib-induced reduction of PD-L1. Knockdown of β-TrCP or inhibition of GSK3 failed to prevent PD-L1 reduction induced by osimertinib. Rather, knockdown of membrane-associated RING-CH 8 (MARCH8) that encodes a membrane-bound E3 ubiquitin ligase rescued osimertinib-induced PD-L1 reduction. Furthermore, manipulation of MARCH8 expression accordingly altered PD-L1 degradation rate. Critically, MARCH8 interacted with PD-L1 through its N-terminal region and also ubiquitinated PD-L1 in cells. Collectively, these results strongly suggest that MARCH8 is a previously undiscovered E3 ubiquitin ligase responsible for PD-L1 degradation including osimertinib-induced PD-L1 degradation, establishing a novel connection between MARCH8 and PD-L1 regulation. Implications: This study has demonstrated a previously undiscovered function of MARCH8 in mediating PD-L1 degradation induced by EGFR inhibitors in EGFR-mutant NSCLC cells, establishing a novel connection between MARCH8 and PD-L1 regulation.
- Published
- 2021
12. Maximizing the value of phase III trials in immuno-oncology: A checklist from the Society for Immunotherapy of Cancer (SITC)
- Author
-
Michael B Atkins, Hamzah Abu-Sbeih, Paolo A Ascierto, Michael R Bishop, Daniel S Chen, Madhav Dhodapkar, Leisha A Emens, Marc S Ernstoff, Robert L Ferris, Tim F Greten, James L Gulley, Roy S Herbst, Rachel W Humphrey, James Larkin, Kim A Margolin, Luca Mazzarella, Suresh S Ramalingam, Meredith M Regan, Brian I Rini, and Mario Sznol
- Subjects
Pharmacology ,Cancer Research ,Clinical Trials as Topic ,Immunology ,Programmed Cell Death 1 Receptor ,Ligands ,Checklist ,Oncology ,Neoplasms ,Molecular Medicine ,Immunology and Allergy ,Animals ,Immunologic Factors ,Immunotherapy ,Immune Checkpoint Inhibitors - Abstract
The broad activity of agents blocking the programmed cell death protein 1 and its ligand (the PD-(L)1 axis) revolutionized oncology, offering long-term benefit to patients and even curative responses for tumors that were once associated with dismal prognosis. However, only a minority of patients experience durable clinical benefit with immune checkpoint inhibitor monotherapy in most disease settings. Spurred by preclinical and correlative studies to understand mechanisms of non-response to the PD-(L)1 antagonists and by combination studies in animal tumor models, many drug development programs were designed to combine anti-PD-(L)1 with a variety of approved and investigational chemotherapies, tumor-targeted therapies, antiangiogenic therapies, and other immunotherapies. Several immunotherapy combinations improved survival outcomes in a variety of indications including melanoma, lung, kidney, and liver cancer, among others. This immunotherapy renaissance, however, has led to many combinations being advanced to late-stage development without definitive predictive biomarkers, limited phase I and phase II data, or clinical trial designs that are not optimized for demonstrating the unique attributes of immune-related antitumor activity—for example, landmark progression-free survival and overall survival. The decision to activate a study at an individual site is investigator-driven, and generalized frameworks to evaluate the potential for phase III trials in immuno-oncology to yield positive data, particularly to increase the number of curative responses or otherwise advance the field have thus far been lacking. To assist in evaluating the potential value to patients and the immunotherapy field of phase III trials, the Society for Immunotherapy of Cancer (SITC) has developed a checklist for investigators, described in this manuscript. Although the checklist focuses on anti-PD-(L)1-based combinations, it may be applied to any regimen in which immune modulation is an important component of the antitumor effect.
- Published
- 2022
13. Osimertinib Versus Comparator EGFR TKI as First-Line Treatment for EGFR-Mutated Advanced NSCLC: FLAURA China, A Randomized Study
- Author
-
M. Saggese, Minhao Fan, Chunling Liu, Andrew Walding, Helong Zhang, Jia Wang, Wei Li, Yong He, X. Huang, Bu-Hai Wang, Qing Zhou, Ying Cheng, and Suresh S. Ramalingam
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,China ,Lung Neoplasms ,medicine.drug_class ,Tyrosine-kinase inhibitor ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,Gefitinib ,Randomized controlled trial ,Double-Blind Method ,law ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Clinical endpoint ,medicine ,Humans ,Pharmacology (medical) ,Osimertinib ,Original Research Article ,Lung cancer ,Protein Kinase Inhibitors ,Aged ,Aged, 80 and over ,Acrylamides ,Aniline Compounds ,business.industry ,Hazard ratio ,Middle Aged ,medicine.disease ,respiratory tract diseases ,ErbB Receptors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Female ,Erlotinib ,business ,medicine.drug - Abstract
Background In the global FLAURA study, first-line osimertinib, a third-generation irreversible tyrosine kinase inhibitor (TKI) of epidermal growth factor receptor (EGFR), significantly improved progression-free survival (PFS) and overall survival (OS) versus comparator EGFR TKIs in patients with EGFR mutation-positive (EGFRm) advanced non-small-cell lung cancer (NSCLC). Objective The FLAURA China study assessed first-line osimertinib in Chinese patients with EGFRm advanced NSCLC (NCT02296125). Methods FLAURA China was a double-blind, randomized, phase III study. Adults from mainland China with previously untreated EGFRm (Exon 19 deletion or L858R) advanced NSCLC were enrolled in the global study or a China-only study under the same protocol; 136 patients were randomized to osimertinib (80 mg once daily [od]; n = 71) or comparator EGFR TKI (gefitinib or erlotinib; all sites selected gefitinib 250 mg od; n = 65). Patients were randomized and allocated to treatment groups by a central computer system. Treatment continued until disease progression, unacceptable toxicity, or withdrawal of consent. The primary endpoint was investigator-assessed PFS; OS was a secondary endpoint. Results All 136 randomized patients were analyzed. Osimertinib extended median PFS by 8.0 months versus comparator EGFR TKI (17.8 vs. 9.8 months; hazard ratio [HR] 0.56; 95% confidence interval [CI] 0.37–0.85). Median OS was 33.1 months in the osimertinib group versus 25.7 months in the comparator group (HR 0.85; 95% CI 0.56–1.29). At 3 years, 20% of patients on osimertinib and 8% on the comparator remained on randomized treatment. Grade 3 or higher adverse events (AEs) were reported in 54 and 28% of patients in the osimertinib and comparator groups, respectively, driven by increased local reporting of laboratory- and disease-related AEs. No new safety signals were identified. Conclusions First-line osimertinib treatment resulted in a clinically meaningful PFS and OS benefit versus comparator EGFR TKI in Chinese patients with EGFRm advanced NSCLC. Safety data were consistent with the known safety profile of osimertinib. Clinical Trial Registration ClinicalTrials.gov NCT02296125, registered 20 November 2014 Supplementary Information The online version contains supplementary material available at 10.1007/s11523-021-00794-6.
- Published
- 2021
14. Downregulation of death receptor 4 is tightly associated with positive response of EGFR mutant lung cancer to EGFR-targeted therapy and improved prognosis
- Author
-
Puyu Shi, Suresh S. Ramalingam, Songqing Fan, Shuo Zhang, Qiming Wang, Yong He, Yixiang Li, Taofeek K. Owonikoko, Shi-Yong Sun, and Zhen Chen
- Subjects
musculoskeletal diseases ,0301 basic medicine ,MAPK/ERK pathway ,Lung Neoplasms ,Down-Regulation ,Medicine (miscellaneous) ,Mice ,03 medical and health sciences ,Transactivation ,0302 clinical medicine ,Downregulation and upregulation ,immune system diseases ,Cell surface receptor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Animals ,Humans ,Osimertinib ,Molecular Targeted Therapy ,death receptor 4 ,Precision Medicine ,skin and connective tissue diseases ,Protein Kinase Inhibitors ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Acrylamides ,Gene knockdown ,Aniline Compounds ,EGFR inhibitors ,Chemistry ,acquired resistance ,apoptosis ,Genes, erbB-1 ,Prognosis ,Xenograft Model Antitumor Assays ,ErbB Receptors ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,030104 developmental biology ,Apoptosis ,Gene Knockdown Techniques ,osimertinib ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Cytokine secretion ,Research Paper - Abstract
Death receptor 4 (DR4), a cell surface receptor, mediates apoptosis or induces inflammatory cytokine secretion upon binding to its ligand depending on cell contexts. Its prognostic impact in lung cancer and connection between EGFR-targeted therapy and DR4 modulation has not been reported and thus was the focus of this study. Methods: Intracellular protein alterations were measured by Western blotting. Cell surface protein was detected with antibody staining and flow cytometry. mRNA expression was monitored with qRT-PCR. Gene transactivation was analyzed with promoter reporter assay. Drug dynamic effects in vivo were evaluated using xenografts. Gene modulations were achieved with gene overexpression and knockdown. Proteins in human archived tissues were stained with immunohistochemistry. Results: EGFR inhibitors (e.g., osimertinib) decreased DR4 levels only in EGFR mutant NSCLC cells and tumors, being tightly associated with induction of apoptosis. This modulation was lost once cells became resistant to these inhibitors. Increased levels of DR4 were detected in cell lines with acquired osimertinib resistance and in NSCLC tissues relapsed from EGFR-targeted therapy. DR4 knockdown induced apoptosis and augmented apoptosis when combined with osimertinib in both sensitive and resistant cell lines, whereas enforced DR4 expression significantly attenuated osimertinib-induced apoptosis. Mechanistically, osimertinib induced MARCH8-mediated DR4 proteasomal degradation and suppressed MEK/ERK/AP-1-dependent DR4 transcription, resulting in DR4 downregulation. Moreover, we found that DR4 positive expression in human lung adenocarcinoma was significantly associated with poor patient survival. Conclusions: Collectively, we suggest that DR4 downregulation is coupled to therapeutic efficacy of EGFR-targeted therapy and predicts improved prognosis, revealing a previously undiscovered connection between EGFR-targeted therapy and DR4 modulation.
- Published
- 2021
15. Inhibition of ACK1 delays and overcomes acquired resistance of EGFR mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib
- Author
-
Nupam P. Mahajan, Jiajia Gu, Suresh S. Ramalingam, Shi-Yong Sun, Guojing Zhang, Taofeek K. Owonikoko, and Luxi Qian
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Cancer Research ,Lung Neoplasms ,Cell ,Mice, Nude ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,Annexin ,Cell Line, Tumor ,Animals ,Medicine ,Osimertinib ,Protein Kinase Inhibitors ,EGFR inhibitors ,Acrylamides ,Aniline Compounds ,Cell growth ,business.industry ,ErbB Receptors ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Resistance, Neoplasm ,Apoptosis ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,business ,Tyrosine kinase - Abstract
Objectives The emergence of acquired resistance to the third generation EGFR inhibitor, osimertinib (AZD9291 or TAGRISSO™), is an unavoidable huge clinical challenge. The involvement of ACK1, a non-receptor tyrosine kinase with an oncogenic function, in regulating cell response to osimertinib has not been investigated and thus is the focus of this study. Material and Methods Drug effects on cell growth were evaluated by measuring cell numbers and colony formation. Apoptosis was monitored with flow cytometry for annexin V-positive cells and Western blotting for protein cleavage. Intracellular protein and mRNA alterations were detected with Western blotting and qRT-PCR, respectively. Drug effects on delaying osimertinib acquired resistance were determined using colony formation in vitro and xenografts in nude mice in vivo, respectively. Cell senescence was assayed by β-galactosidase staining. Results Inhibition of ACK1 with the novel ACK1 inhibitor, (R)-9b synergized with osimertinib in inhibiting the growth of EGFR mutant NSCLC cell lines. Similar results were also generated with ACK1 gene knockdown. The combination of osimertinib and (R)-9b enhanced induction of apoptosis. In both in vitro and in vivo long-term resistance delay assays, the combination of (R)-9b and osimertinib clearly delayed the emergence of osimertinib-resistance. Further, the (R)-9b and osimertinib combination was also effective in inhibiting the growth of EGFR mutant NSCLC cell lines with acquired resistance to osimertinib, which possess elevated levels of ACK1, and the growth of osimertinib-resistant tumors in vivo. In some resistant cell lines, the combinations induced senescence in addition to induction of apoptosis. Conclusions These novel findings suggest that ACK1 inhibition might be a potential and innovative strategy for delaying and overcoming osimertinb acquired resistance.
- Published
- 2020
16. Phase Ib Study of Chemoprevention with Green Tea Polyphenon E and Erlotinib in Patients with Advanced Premalignant Lesions (APL) of the Head and Neck
- Author
-
Zhengjia Chen, Fadlo R. Khuri, Sreenivas Nannapaneni, Dong M. Shin, Qiuying Shi, Conor E. Steuer, Jonathan J. Beitler, Mark W. El-Deiry, Adam M. Klein, Mihir R. Patel, Taofeek K. Owonikoko, Yuan Liu, Amy Y. Chen, Steven M. Roser, Zhuo Georgia Chen, Suresh S. Ramalingam, and Nabil F. Saba
- Subjects
0301 basic medicine ,Larynx ,Cancer Research ,medicine.medical_specialty ,business.industry ,Carcinoma in situ ,Head and neck cancer ,Polyphenon E ,medicine.disease ,Rash ,Gastroenterology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Internal medicine ,Toxicity ,medicine ,Biomarker (medicine) ,Erlotinib ,medicine.symptom ,business ,medicine.drug - Abstract
Purpose: On the basis of synergistic effects between green tea polyphenon E (PPE) and EGFR–tyrosine kinase inhibitor in preclinical studies, we conducted a phase Ib study of the PPE and erlotinib combination in patients with advanced premalignant lesions (APL) of the oral cavity and larynx. Patients and Methods: Patients were treated with a fixed dose of PPE (200 mg three times a day) and dose escalation of erlotinib (50, 75, 100 mg daily) for 6 months with tissue biopsy at baseline and 6 months. Primary endpoints were safety and toxicity; secondary endpoints were evaluation of pathologic response, cancer-free survival (CFS), overall survival (OS), and biomarker modulation. Results: Among 21 enrolled patients, 19 began treatment and 17 completed 6 months of treatment with PPE and erlotinib. Main characteristics of treated patients: 15 severe dysplasia or carcinoma in situ and 17 oral cavity. Only skin rash was associated with dose-limiting toxicity and MTD. Recommended doses for phase II studies are PPE 600 mg daily plus erlotinib 100 mg daily for 6 months. Pathologic responses in 17 evaluable patients: pathologic complete response (47%) and pathologic partial response (18%). The 5-year CFS and OS were 66.3% and 93%, respectively. Among tested biomarkers, only phosphorylated ERK was correlated with response to treatment. Conclusions: Treatment with PPE and erlotinib combination was well tolerated in patients with APLs of the head and neck, and showed a high rate of pathologic response with excellent CFS. This combination deserves further investigation for the chemoprevention and/or prevention of second primary tumors in early-stage head and neck cancer.
- Published
- 2020
17. Phase II Study of Immunotherapy With Tecemotide and Bevacizumab After Chemoradiation in Patients With Unresectable Stage III Non-Squamous Non–Small-Cell Lung Cancer (NS-NSCLC): A Trial of the ECOG-ACRIN Cancer Research Group (E6508)
- Author
-
Anil Shanker, Millie Das, Melissa Lynne Johnson, David P. Carbone, Henry N. Wagner, Joan H. Schiller, Christopher S.R. Dakhil, Leora Horn, Maria Teresa P. de Aquino, Suresh S. Ramalingam, David E. Gerber, Ju Whei Lee, Mohammed Ali Al-Nsour, Jyoti D. Patel, and Jane Jijun Liu
- Subjects
Male ,0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,genetic structures ,Paclitaxel ,Bevacizumab ,Phases of clinical research ,Adenocarcinoma of Lung ,Cancer Vaccines ,Article ,Carboplatin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Membrane Glycoproteins ,business.industry ,Chemoradiotherapy ,Middle Aged ,Prognosis ,Survival Rate ,Regimen ,030104 developmental biology ,chemistry ,Tolerability ,030220 oncology & carcinogenesis ,Carcinoma, Large Cell ,Tecemotide ,Female ,Immunotherapy ,business ,Follow-Up Studies ,medicine.drug - Abstract
Introduction Although chemoradiotherapy (CRT) is the standard of care for patients with unresectable stage III non–small-cell lung cancer (LA-NSCLC), most patients relapse. Tecemotide is a MUC1 antigen-specific cancer immunotherapy vaccine. Bevacizumab improves survival in advanced nonsquamous (NS)-NSCLC and has a role in immune modulation. This phase II trial tested the combination of tecemotide and bevacizumab following CRT in patients with LA-NSCLC. Patients and Methods Subjects with stage III NS-NSCLC suitable for CRT received carboplatin/paclitaxel weekly + 66 Gy followed by 2 cycles of consolidation carboplatin/paclitaxel ≤ 4 weeks of completion of CRT (Step 1). Patients with partial response/stable disease after consolidation therapy were registered onto step 2, which was 6 weekly tecemotide injections followed by every 6 weekly injections and bevacizumab every 3 weeks for up to 34 doses. The primary endpoint was to determine the safety of this regimen. Results Seventy patients were enrolled; 68 patients (median age, 63 years; 56% male; 57% stage IIIA) initiated therapy, but only 39 patients completed CRT and consolidation therapy per protocol, primarily owing to disease progression or toxicity. Thirty-three patients (median age, 61 years; 58% male; 61% stage IIIA) were registered to step 2 (tecemotide + bevacizumab). The median number of step 2 cycles received was 11 (range, 2-25). Step 2 worst toxicity included grade 3, N = 9; grade 4, N = 1; and grade 5, N = 1. Grade 5 toxicity in step 2 was esophageal perforation attributed to bevacizumab. Among the treated and eligible patients (n = 32) who were treated on step 2, the median overall survival was 42.7 months (95% confidence interval, 21.7-63.3 months), and the median progression-free survival was 14.9 months (95% confidence interval, 11.0-20.9 months) from step 1 registration. Conclusions This cooperative group trial met its endpoint, demonstrating tolerability of bevacizumab + tecemotide after CRT and consolidation. In this selected group of patients, the median progression-free survival and overall survival are encouraging. Given that consolidation immunotherapy is now a standard of care following CRT in patients with LA-NSCLC, these results support a role for continued investigation of antiangiogenic and immunotherapy combinations in LA-NSCLC.
- Published
- 2020
18. Efficacy and safety of immune checkpoint blockade in self‐identified Black patients with advanced non–small cell lung cancer
- Author
-
Tyler Beardslee, Yuan Liu, Suresh S. Ramalingam, Subir Goyal, Taofeek K. Owonikoko, Levani Odikadze, Harpaul S. Gill, Anne Engelhart, Manoj K. Mishra, Zhengjia Chen, Bassel Nazha, and Jennifer W Carlisle
- Subjects
Adult ,Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Georgia ,Lung Neoplasms ,Multivariate analysis ,Kaplan-Meier Estimate ,Pembrolizumab ,Article ,03 medical and health sciences ,0302 clinical medicine ,Atezolizumab ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Lung cancer ,Adverse effect ,Immune Checkpoint Inhibitors ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Middle Aged ,medicine.disease ,Blockade ,Black or African American ,Treatment Outcome ,Tolerability ,030220 oncology & carcinogenesis ,Multivariate Analysis ,Female ,Nivolumab ,business - Abstract
Background To the authors' knowledge, race-based differences in efficacy for the treatment of patients with advanced non-small cell lung cancer (NSCLC) have not been studied to date due to the underrepresentation of patients of minority backgrounds in pivotal trials. In the current study, the authors examined real-world differences in outcome in a diverse patient population. Methods The authors retrospectively analyzed the clinical outcomes of patients with advanced NSCLC who were treated with single-agent immune checkpoint blockade (ICB) between 2013 and July 2018 at Winship Cancer Institute of Emory University in Atlanta, Georgia. Primary efficacy comparison between Black patients and White patients was performed using bivariate and multivariate analyses for overall survival (OS) and progression-free survival (PFS). Results Data from 257 patients were analyzed. The median age of the patients was 69 years; 50.6% of the patients were female, 63.4% were White, 29.5% were Black, and 7.1% of the patients were of "other" race. ICB was the first-line treatment in 51 patients (19.9%), the second-line treatment in 161 patients (62.6%), and the third-line treatment in 33 patients (12.9%). The most commonly used agents were nivolumab (49.0%), pembrolizumab (25.2%), and atezolizumab (21.3%). No differences with regard to OS (P = .839) and PFS (P = .235) were noted between Black and White patients. The sample overall response rate was 20.6% (15.2% in Black patients and 23.1% in White patients). No differences with regard to OS (P = .081) and PFS (P = .176) were observed between female and male patients. The rate of immune-related adverse events was found to be similar in Black and White patients (20.0% vs 29.9%; P = .148). On multivariate analysis, race was not found to be significantly associated with OS or PFS. Conclusions Real-world analysis of the authors' institutional experience demonstrated similar efficacy and tolerability of ICB in Black versus White patients with advanced NSCLC. Larger multi-institutional studies including other US minority populations would make the findings of the current study more generalizable.
- Published
- 2020
19. Phase 1 safety and pharmacodynamic study of lenalidomide combined with everolimus in patients with advanced solid malignancies with efficacy signal in adenoid cystic carcinoma
- Author
-
Christina Wu, Chao Zhang, Mehmet Asim Bilen, Walid L. Shaib, Sagar Lonial, Bassel F. El-Rayes, Wayne Harris, Edmund K. Waller, Fadlo R. Khuri, David H. Lawson, Mohammad S. Hossain, R. Donald Harvey, Colleen Lewis, Suresh S. Ramalingam, Olatunji B. Alese, Conor E. Steuer, Zhengjia Chen, Taofeek K. Owonikoko, and Bradley C. Carthon
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Adenoid cystic carcinoma ,Drug development ,Neutropenia ,Article ,03 medical and health sciences ,0302 clinical medicine ,Targeted therapies ,Internal medicine ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Everolimus ,Adverse effect ,Lenalidomide ,030304 developmental biology ,0303 health sciences ,business.industry ,Middle Aged ,medicine.disease ,Rash ,Carcinoma, Adenoid Cystic ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Cytokines ,Female ,medicine.symptom ,business ,Progressive disease ,medicine.drug - Abstract
Background Purpose: The combination of a mammalian target of rapamycin inhibitor and lenalidomide showed enhanced preclinical cytotoxicity. We conducted a phase 1 study in advanced solid tumour patients to assess safety, efficacy and pharmacodynamic (PD) outcomes. Methods We employed a 3+3 dose escalation design to establish the safety and recommended phase 2 doses (RP2D) of daily everolimus and lenalidomide in patients with advanced solid tumours. The starting doses were 5 and 10 mg, respectively, with planned escalation to maximum single-agent doses of 10 and 25 mg in the absence of dose-limiting toxicity. PD endpoints of lymphocyte subsets and immune cytokines were assessed in peripheral blood using multiparameter flow cytometry and LUMINEX assay. Efficacy was evaluated by cross-sectional imaging after every two cycles of treatment. Results The study enrolled 44 patients, median age of 58 years and 28 males (63.6%). The RP2D was established as 10 and 25 mg daily continuously for everolimus and lenalidomide. Common (>5%) grade ≥3 adverse events included rash (19%), neutropenia (19%), hypokalaemia (11%) and fatigue (9%). Best efficacy outcomes in 36 evaluable patients were partial response in 5 (13.8%), stable disease in 24 (55.8%) and progressive disease in 7 (19.4%) patients. PD assessment revealed significant association of cytokine levels (interleukin-2 (IL2), IL21 and IL17), baseline activated and total CD8+ lymphocytes and change in B cell lymphocytes and activated NK cells with clinical benefit. Conclusions The study demonstrated the safety of everolimus and lenalidomide with promising efficacy signal in thyroid and adenoid cystic cancers. Clinical Trial Registration NCT01218555
- Published
- 2020
20. MEK or ERK inhibition effectively abrogates emergence of acquired osimertinib resistance in the treatment of epidermal growth factor receptor–mutant lung cancers
- Author
-
Jiajia Gu, Guojing Zhang, Puyu Shi, Taofeek K. Owonikoko, Weilong Yao, Suresh S. Ramalingam, and Shi-Yong Sun
- Subjects
MAPK/ERK pathway ,Cancer Research ,biology ,medicine.diagnostic_test ,business.industry ,Cell ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Apoptosis ,In vivo ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Medicine ,Osimertinib ,030212 general & internal medicine ,Epidermal growth factor receptor ,business ,EGFR inhibitors - Abstract
Background The majority of patients with non-small cell lung cancer (NSCLC) harboring activating epidermal growth factor receptor (EGFR) mutations respond well to osimertinib (AZD9291), a third-generation, mutation-selective EGFR inhibitor. The current study focuses on determining whether targeting MEK/ERK signaling prevents or delays the development of acquired resistance to osimertinib. Methods Drug effects on cell survival were determined by measuring cell number alterations. Apoptosis was assessed with flow cytometry for the detection of annexin V-positive cells and with Western blotting for protein cleavage. Alterations of proteins in cells were detected with Western blotting. Drug effects on delaying the emergence of osimertinib resistance were evaluated with colony formation in vitro and xenografts in nude mice in vivo. Results Osimertinib combined with an MEK or ERK inhibitor synergistically decreased cell survival with enhanced induction of apoptosis in EGFR-mutant NSCLC cells but not in EGFR wild-type NSCLC cells. These combinations were also very effective in killing cell clones with primary intrinsic resistance to osimertinib. Continuous and intermittent pharmacologic inhibition of MEK/ERK signaling delayed the emergence of osimertinib resistance both in vitro and in vivo. Conclusions These results provide strong preclinical evidence in support of targeting MEK/ERK signaling as a strategy for delaying or preventing acquired resistance to osimertinib in the clinic to improve the long-term therapeutic efficacy of osimertinib. From a clinical standpoint, the data support the evaluation of an intermittent treatment schedule of osimertinib in combination with an MEK or ERK inhibitor in patients with EGFR-mutated NSCLC.
- Published
- 2020
21. A Phase I Study of Safety, Pharmacokinetics, and Pharmacodynamics of Concurrent Everolimus and Buparlisib Treatment in Advanced Solid Tumors
- Author
-
Suresh S. Ramalingam, Conor E. Steuer, Mehmet Asim Bilen, Hannah Collins, Sagar Lonial, Zhengjia Chen, Bradley C. Carthon, Mehmet Akce, Olatunji B. Alese, Taofeek K. Owonikoko, Christina Wu, Ragini R Kudchagkar, David H. Lawson, R. Donald Harvey, Colleen Lewis, Chao Zhang, Bassel F. El-Rayes, Wayne Harris, Walid L. Shaib, Gabriel Sica, and Fadlo R. Khuri
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Maximum Tolerated Dose ,Nausea ,Morpholines ,Buparlisib ,Aminopyridines ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Pharmacokinetics ,Neoplasms ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Mucositis ,Humans ,Tissue Distribution ,Everolimus ,Survival rate ,Aged ,business.industry ,Middle Aged ,Prognosis ,medicine.disease ,Survival Rate ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Pharmacodynamics ,Female ,medicine.symptom ,business ,Progressive disease ,Follow-Up Studies ,medicine.drug - Abstract
Purpose: Concurrent inhibition of mTOR and PI3K led to improved efficacy in preclinical models and provided the rationale for this phase I study of everolimus and buparlisib (BKM120) in patients with advanced solid tumor. Patients and Methods: We used the Bayesian Escalation with Overdose Control design to test escalating doses of everolimus (5 or 10 mg) and buparlisib (20, 40, 60, 80, and 100 mg) in eligible patients. Pharmacokinetic assessment was conducted using blood samples collected on cycle 1, days 8 and 15. Pharmacodynamic impact on mTOR/PI3K pathway modulation evaluated in paired skin biopsies collected at baseline and end of cycle 1. Results: We enrolled 43 patients, median age of 63 (range, 39–78) years; 25 (58.1%) females, 35 (81.4%) Caucasians, and 8 (18.6%) Blacks. The most frequent toxicities were hyperglycemia, diarrhea, nausea, fatigue, and aspartate aminotransferase elevation. Dose-limiting toxicities observed in 7 patients were fatigue (3), hyperglycemia (2), mucositis (1), acute kidney injury (1), and urinary tract infection (1). The recommended phase II dose (RP2D) for the combination was established as everolimus (5 mg) and buparlisib (60 mg). The best response in 27 evaluable patients was progressive disease and stable disease in 3 (11%) and 24 (89%), respectively. The median progression-free survival and overall survival were 2.7 (1.8–4.2) and 9 (6.4–13.2) months. Steady-state pharmacokinetic analysis showed dose-normalized maximum concentrations and AUC values for everolimus and buparlisib in combination to be comparable with single-agent pharmacokinetic. Conclusions: The combination of everolimus and buparlisib is safe and well-tolerated at the RP2D of 5 and 60 mg on a continuous daily schedule.
- Published
- 2020
22. Prognostic significance of an invasive leader cell–derived mutation cluster on chromosome 16q
- Author
-
Bhakti Dwivedi, Adam I. Marcus, Suresh S. Ramalingam, Manali Rupji, Brian Pedro, Paula M. Vertino, Jeanne Kowalski, Jessica Konen, and Taofeek K. Owonikoko
- Subjects
Adult ,Male ,Cancer Research ,Cell ,Genomics ,Article ,Metastasis ,03 medical and health sciences ,0302 clinical medicine ,Genotype-phenotype distinction ,Carcinoma, Non-Small-Cell Lung ,Biomarkers, Tumor ,medicine ,Humans ,Cell Lineage ,Neoplasm Invasiveness ,030212 general & internal medicine ,Lung cancer ,Gene ,Aged ,Aged, 80 and over ,Sequence Analysis, RNA ,business.industry ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,Multigene Family ,030220 oncology & carcinogenesis ,Hepatocellular carcinoma ,Mutation ,Cancer research ,Adenocarcinoma ,Female ,business ,Chromosomes, Human, Pair 16 - Abstract
Background Intratumoral heterogeneity is defined by subpopulations with varying genotypes and phenotypes. Specialized, highly invasive leader cells and less invasive follower cells are phenotypically distinct subpopulations that cooperate during collective cancer invasion. Because leader cells are a rare subpopulation that would be missed by bulk sequencing, a novel image-guided genomics platform was used to precisely select this subpopulation. This study identified a novel leader cell mutation signature and tested its ability to predict prognosis in non-small cell lung cancer (NSCLC) patient cohorts. Methods Spatiotemporal genomic and cellular analysis was used to isolate and perform RNA sequencing on leader and follower populations from the H1299 NSCLC cell line, and it revealed a leader-specific mutation cluster on chromosome 16q. Genomic data from patients with lung squamous cell carcinoma (LUSC; n = 475) and lung adenocarcinoma (LUAD; n = 501) from The Cancer Genome Atlas were stratified by 16q mutation cluster (16qMC) status (16qMC+ vs 16qMC-) and compared for overall survival (OS), progression-free survival (PFS), and gene set enrichment analysis (GSEA). Results Poorer OS, poorer PFS, or both were found across all stages and among early-stage patients with 16qMC+ tumors within the LUSC and LUAD cohorts. GSEA revealed 16qMC+ tumors to be enriched for the expression of metastasis- and survival-associated gene sets. Conclusions This represents the first leader cell mutation signature identified in patients and has the potential to better stratify high-risk NSCLC and ultimately improve patient outcomes.
- Published
- 2020
23. Overcoming acquired resistance of EGFR‐mutant NSCLC cells to the third generation EGFR inhibitor, osimertinib, with the natural product honokiol
- Author
-
Guoqing Qian, Jack L. Arbiser, Songqing Fan, Shi-Yong Sun, Hongjing Zang, Taofeek K. Owonikoko, and Suresh S. Ramalingam
- Subjects
0301 basic medicine ,Honokiol ,Cancer Research ,Lung Neoplasms ,Cell ,honokiol ,chemistry.chemical_compound ,T790M ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Osimertinib ,Epidermal growth factor receptor ,Research Articles ,EGFR inhibitors ,Aniline Compounds ,biology ,Chemistry ,apoptosis ,Drug Synergism ,General Medicine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,ErbB Receptors ,medicine.anatomical_structure ,Oncology ,osimertinib ,030220 oncology & carcinogenesis ,Molecular Medicine ,Research Article ,EGFR ,Mice, Nude ,lcsh:RC254-282 ,Lignans ,03 medical and health sciences ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,Lung cancer ,Protein Kinase Inhibitors ,Acrylamides ,Biphenyl Compounds ,acquired resistance ,medicine.disease ,Antineoplastic Agents, Phytogenic ,lung cancer ,030104 developmental biology ,Drug Resistance, Neoplasm ,Apoptosis ,Mutation ,Cancer research ,biology.protein - Abstract
The development of acquired resistance to osimertinib (Osim) (AZD9291 or TAGRISSOTM), an FDA‐approved third‐generation epidermal growth factor receptor (EGFR) inhibitor for the treatment of EGFR‐mutant nonsmall cell lung cancer (NSCLC), limits the long‐term benefits for patients. Thus, effective treatment options are urgently needed. To this end, we explored whether honokiol (HNK), a natural product with potential antitumor activity, may be used to overcome Osim resistance. The combination of HNK and Osim synergistically decreased the survival of several Osim ‐resistant cell lines with enhanced effects on inhibiting cell colony formation and growth and on inducing apoptosis. This combination also showed greater growth suppression of Osim‐resistant xenograft tumors including those with 19del, T790M, and C797S triple mutations in nude mice. Mechanistically, the augmented induction of apoptosis by the combination is largely due to enhanced Mcl‐1 reduction through facilitating its degradation. A synthetic HNK derivative exerted similar effects with greater efficacy. Our findings warrant further study of HNK and its derivatives in overcoming Osim resistance in the clinic., The development of acquired resistance to osimertinib (Osim), an FDA‐approved third‐generation epidermal growth factor receptor (EGFR) inhibitor for the treatment of EGFR‐mutant nonsmall cell lung cancer, limits the long‐term benefits for patients. The natural product, honokiol, when combined with Osim, augmented induction of apoptosis in different Osim ‐resistant cell lines primarily through enhancing Mcl‐1 degradation and effectively inhibited the growth of Osim‐resistant tumors.
- Published
- 2020
24. Randomized Phase III Study of Ganetespib, a Heat Shock Protein 90 Inhibitor, With Docetaxel Versus Docetaxel in Advanced Non–Small-Cell Lung Cancer (GALAXY-2)
- Author
-
Michael Schenker, Vladimir Kovcin, Florentina Teofilovici, Rodryg Ramlau, Dariusz M. Kowalski, Rathi N. Pillai, Tudor-Eliade Ciuleanu, V. Vukovic, Ilker Yalcin, Dean A. Fennell, and Suresh S. Ramalingam
- Subjects
Adult ,Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Ganetespib ,Docetaxel ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Heat shock protein ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,HSP90 Heat-Shock Proteins ,Lung cancer ,Aged ,Aged, 80 and over ,Salvage Therapy ,Antitumor activity ,business.industry ,Middle Aged ,Triazoles ,medicine.disease ,Progression-Free Survival ,Treatment Outcome ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,Non small cell ,business ,medicine.drug - Abstract
PURPOSE Ganetespib, a highly potent heat shock protein 90 inhibitor, blocks multiple oncogenic pathways, resulting in antitumor activity. We evaluated the combination of ganetespib and docetaxel for second-line therapy of patients with advanced adenocarcinoma of the lung. PATIENTS AND METHODS In this international phase III trial, patients with stage IIIB or IV adenocarcinoma diagnosed > 6 months before study entry and 1 prior systemic therapy were randomly assigned (1:1) to ganetespib 150 mg/m2 on days 1 and 15 with docetaxel 75 mg/m2 on day 1 of a 21-day cycle or to docetaxel alone. The primary end point was overall survival (OS). RESULTS Of 677 enrolled patients, 335 were randomly assigned to ganetespib and docetaxel and 337 were assigned to docetaxel. The trial was stopped early as a result of futility at a planned interim analysis. The median OS time was 10.9 months (95% CI, 9.0 to 12.3 months) in the ganetespib and docetaxel arm compared with 10.5 months (95% CI, 8.6 to 12.2 months) in docetaxel arm (hazard ratio [HR], 1.11; 95% CI, 0.899 to 1.372; P = .329). Median progression-free survival was 4.2 months in the ganetespib and docetaxel arm and 4.3 months in the docetaxel arm (HR, 1.16; 95% CI, 0.96 to 1.403; P = .119). The addition of ganetespib did not improve outcomes compared with docetaxel alone for any secondary end point, including survival in the elevated lactate dehydrogenase or EGFR and ALK wild-type populations. The most common grade 3 or 4 adverse event in both arms was neutropenia (30.9% with ganetespib and docetaxel v 25% with docetaxel). CONCLUSION The addition of ganetespib to docetaxel did not result in improved survival for salvage therapy of patients with advanced-stage lung adenocarcinoma.
- Published
- 2020
25. Overcoming acquired resistance of epidermal growth factor receptor‐mutant non–small cell lung cancer cells to osimertinib by combining osimertinib with the histone deacetylase inhibitor panobinostat (LBH589)
- Author
-
Guoqing Qian, Hongjing Zang, Taofeek K. Owonikoko, Dan Zong, Songqing Fan, Suresh S. Ramalingam, and Shi-Yong Sun
- Subjects
Cancer Research ,Lung Neoplasms ,medicine.drug_class ,Cell ,Antineoplastic Agents ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Epidermal growth factor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Panobinostat ,Humans ,Medicine ,Osimertinib ,030212 general & internal medicine ,Epidermal growth factor receptor ,Protein Kinase Inhibitors ,Acrylamides ,Aniline Compounds ,biology ,business.industry ,Cell growth ,Histone deacetylase inhibitor ,ErbB Receptors ,Histone Deacetylase Inhibitors ,medicine.anatomical_structure ,Oncology ,chemistry ,Drug Resistance, Neoplasm ,Cell culture ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,biology.protein ,Drug Therapy, Combination ,business - Abstract
BACKGROUND The major clinical obstacle that limits the long-term benefits of treatment with osimertinib (AZD9291) in patients with epidermal growth factor receptor-mutant non-small cell lung cancer is the development of acquired resistance. Therefore, effective strategies that can overcome acquired resistance to osimertinib are urgently needed. The authors' current efforts in this direction have identified LBH589 (panobinostat), a clinically used histone deacetylase inhibitor, as a potential agent in overcoming osimertinib resistance. METHODS Cell growth and apoptosis in vitro were evaluated by measuring cell numbers and colony formation and by detecting annexin V-positive cells and protein cleavage, respectively. Drug effects on tumor growth in vivo were assessed with xenografts in nude mice. Alterations of tested proteins in cells were monitored with Western blot analysis. Gene knockout was achieved using the CRISPR/Cas9 technique. RESULTS The combination of LBH589 and osimertinib synergistically decreased the survival of different osimertinib-resistant cell lines, including those harboring C797S mutations, with greater inhibition of cell colony formation and growth. The combination enhanced the induction of apoptosis in osimertinib-resistant cells. Importantly, the combination effectively inhibited the growth of osimertinib-resistant xenograft tumors in nude mice. Mechanistically, the combination of LBH589 and osimertinib enhanced the elevation of Bim in osimertinib-resistant cells. Knockout of Bim in osimertinib-resistant cells substantially attenuated or abolished apoptosis enhanced by the LBH589 and osimertinib combination. These results collectively support a critical role of Bim elevation in the induction of apoptosis of osimertinib-resistant cells for this combination. CONCLUSIONS The current findings provide strong preclinical evidence in support of the potential for LBH589 to overcome osimertinib resistance in the clinic.
- Published
- 2020
26. Trimodality Therapy in the Treatment of Stage III N2-Positive Non-Small Cell Lung Cancer: A National Cancer Database Analysis
- Author
-
Walter J. Curran, Conor E. Steuer, Theresa W. Gillespie, Seth D. Force, Madhusmita Behera, Chandra P. Belani, Kristin Higgins, Suresh S. Ramalingam, Fadlo R. Khuri, Rathi N. Pillai, Yuan Liu, Suchita Pakkala, Felix G. Fernandez, and Chao Fu
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,030204 cardiovascular system & hematology ,03 medical and health sciences ,Pneumonectomy ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Humans ,Stage (cooking) ,Lung cancer ,Aged ,Neoplasm Staging ,Retrospective Studies ,Proportional hazards model ,business.industry ,Lung Cancer ,Hazard ratio ,Cancer ,Odds ratio ,medicine.disease ,Combined Modality Therapy ,Radiation therapy ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,business - Abstract
Background Significant controversy remains regarding the care of patients with clinical stage III (N2-positive) NSCLC. Although multimodality therapy is effective, the roles of surgery, chemotherapy, and radiotherapy are not fully defined and the optimal treatment approach is not firmly established. We analyzed outcomes and predictors associated with trimodality therapy (TT) in the National Cancer Database. Materials and Methods The NCDB was queried from 2004 to 2014 for patients with NSCLC diagnosed with stage III (N2) disease and treated with chemotherapy and radiation (CRT). Three cohorts of patients were studied: CRT only/no surgery (NS), CRT plus lobectomy (LT), and CRT plus pneumonectomy (PT). The univariate and multivariable analyses (MVA) were conducted using Cox proportional hazards model and log-rank tests. Results A total of 29,754 patients were included in this analysis: NS 90.1%, LT 8.4%, and PT 1.5%. Patient characteristics: median age 66 years; male 56% and white 85%. Patients treated at academic centers were more likely to receive TT compared with those treated at community centers (odds ratio: 1.85 [1.53–2.23]; p < .001). On MVA, patients that received TT were associated with better survival than those that received only CRT (hazard ratio: 0.59 [0.55–0.62]; p < .001). The LT group was associated with significantly better survival than the PT and NS groups (median survival: 62.8 months vs. 51.8 months vs. 34.2 months, respectively). In patients with more than two nodes involved, PT was associated with worse survival than LT and NS (median survival: 51.4 months in LT and 39 months in NS vs. 37 months in PT). The 30-day and 90-day mortality rates were found to be significantly higher in PT patients than in LT. Conclusion TT was used in less than 10% of patients with stage III N2 disease, suggesting high degree of patient selection. In this selected group, TT was associated with favorable outcomes relative to CRT alone. Implications for Practice This analysis demonstrates that trimodality therapy could benefit a selected subset of patients with stage III (N2) disease. This plan should be considered as a treatment option following patient evaluation in a multidisciplinary setting in experienced medical centers with the needed expertise.
- Published
- 2020
27. Correction to: Osimertinib for EGFR-Mutant Non-Small-Cell Lung Cancer Central Nervous System Metastases: Current Evidence and Future Perspectives on Therapeutic Strategies
- Author
-
Sanjay Popat, Myung-Ju Ahn, Simon Ekman, Natasha B. Leighl, Suresh S. Ramalingam, Thanyanan Reungwetwattana, Shankar Siva, Masahiro Tsuboi, Yi-Long Wu, and James Chih-Hsin Yang
- Subjects
Cancer Research ,Oncology ,Pharmacology (medical) - Published
- 2023
28. Mcl-1 Interacts with Akt to Promote Lung Cancer Progression
- Author
-
Chao Zhang, Keqiang Ye, Walter J. Curran, Gabriel Sica, Madhusmita Behera, Xingming Deng, Guo Chen, Dongkyoo Park, Taofeek K. Owonikoko, Zhengjia Chen, Suresh S. Ramalingam, and Andrew T. Magis
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Antineoplastic Agents ,Kaplan-Meier Estimate ,Article ,Small hairpin RNA ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Lung cancer ,Lung ,Protein kinase B ,Kinase ,Chemistry ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Molecular Docking Simulation ,Pleckstrin homology domain ,030104 developmental biology ,Oncology ,Protein kinase domain ,Tissue Array Analysis ,030220 oncology & carcinogenesis ,Cancer cell ,Disease Progression ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,CRISPR-Cas Systems ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Follow-Up Studies ,Protein Binding ,Signal Transduction - Abstract
Mcl-1 is a unique antiapoptotic Bcl2 family protein that functions as a gatekeeper in manipulating apoptosis and survival in cancer cells. Akt is an oncogenic kinase that regulates multiple cellular functions and its activity is significantly elevated in human cancers. Here we discovered a cross-talk between Mcl-1 and Akt in promoting lung cancer cell growth. Depletion of endogenous Mcl-1 from human lung cancer cells using CRISPR/Cas9 or Mcl-1 shRNA significantly decreased Akt activity, leading to suppression of lung cancer cell growth in vitro and in xenografts. Mechanistically, Mcl-1 directly interacted via its PEST domain with Akt at the pleckstrin homology (PH) domain. It is known that the interactions between the PH domain and kinase domain (KD) are important for maintaining Akt in an inactive state. The binding of Mcl-1/PH domain disrupted intramolecular PH/KD interactions to activate Akt. Intriguingly, Mcl-1 expression correlated with Akt activity in tumor tissues from patients with non–small cell lung cancer. Using the Mcl-1–binding PH domain of Akt as a docking site, we identified a novel small molecule, PH-687, that directly targets the PH domain and disrupts Mcl-1/Akt binding, leading to suppression of Akt activity and growth inhibition of lung cancer in vitro and in vivo. By targeting the Mcl-1/Akt interaction, this mechanism-driven agent provides a highly attractive strategy for the treatment of lung cancer. Significance: These findings indicate that targeting Mcl-1/Akt interaction by employing small molecules such as PH-687 represents a potentially new and effective strategy for cancer treatment.
- Published
- 2019
29. Combined Effect of Sarcopenia and Systemic Inflammation on Survival in Patients with Advanced Stage Cancer Treated with Immunotherapy
- Author
-
David H. Lawson, Haydn T. Kissick, R. Donald Harvey, Jacqueline T. Brown, Colleen Lewis, Olatunji B. Alese, Ragini R. Kudchadkar, Bassel F. El-Rayes, Mehmet Asim Bilen, Walid L. Shaib, Christina Wu, Amir Ishaq Khan, Yuan Liu, Julie M. Shabto, Hannah Collins, Rathi N. Pillai, Suresh S. Ramalingam, Milton Williams, Alexandra Speak, Viraj A. Master, Dylan J. Martini, Conor E. Steuer, Bradley C. Carthon, Mehmet Akce, and Taofeek K. Owonikoko
- Subjects
Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Sarcopenia ,Systemic inflammation ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Neoplasms ,Medicine ,Humans ,Lymphocyte Count ,Letters to the Editor ,Risk stratification ,030304 developmental biology ,Retrospective Studies ,Inflammation ,0303 health sciences ,business.industry ,Melanoma ,Hazard ratio ,Cancer ,medicine.disease ,Prognosis ,Immuno‐Oncology ,Confidence interval ,3. Good health ,Clinical trial ,030220 oncology & carcinogenesis ,Cohort ,Female ,Immunotherapy ,medicine.symptom ,business ,Biomarkers - Abstract
Background Sarcopenia and inflammation have been associated with poor survival in patients with cancer. We explored the combined effects of these variables on survival in patients with cancer treated with immunotherapy. Methods We performed a retrospective review of 90 patients enrolled on immunotherapy‐based phase I clinical trials at Emory University from 2009 to 2017. Baseline neutrophil‐to‐lymphocyte ratio, monocyte‐to‐lymphocyte ratio, and platelet‐to‐lymphocyte ratio (PLR) were used as surrogates of inflammation. The skeletal muscle index (SMI) was derived from the skeletal muscle density calculated from baseline abdominal computed tomography images. Optimal cutoffs for continuous inflammation biomarkers and SMI were determined by bias‐adjusted log‐rank test. A four‐level risk stratification was used to create low‐risk (PLR, The interaction between chronic inflammation and body composition is particularly important in the era of immunotherapy, considering that immune checkpoint inhibitors rely on the host immune system for their efficacy. This article reports on the combined effects of inflammation and sarcopenia on clinical outcomes in patients with solid tumors treated with immunotherapy‐based regimens.
- Published
- 2019
30. Encorafenib plus binimetinib in patients with BRAF(V)(600)-mutant non-small cell lung cancer
- Author
-
Gregory J Riely, Myung-Ju Ahn, Enriqueta Felip, Suresh S Ramalingam, Egbert F Smit, Anne S Tsao, Ann Alcasid, Tiziana Usari, Paul S Wissel, Keith D Wilner, Bruce E Johnson, Institut Català de la Salut, [Riely GJ] Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA. [Ahn MJ] Department of Hematology and Oncology, Samsung Medical Center, Seoul, Republic of Korea. [Felip E] Servei d’Oncologia Mèdica, Vall d’Hebron Hospital Universitari, Barcelona, Spain. Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain. [Ramalingam SS] Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, USA. [Smit EF] Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands. [Tsao AS] Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, USA, and Vall d'Hebron Barcelona Hospital Campus
- Subjects
safety ,Cancer Research ,Mutació (Biologia) ,efficacy ,Otros calificadores::Otros calificadores::/farmacoterapia [Otros calificadores] ,clinical trial ,encorafenib ,General Medicine ,Neoplasms::Neoplasms by Site::Thoracic Neoplasms::Respiratory Tract Neoplasms::Lung Neoplasms::Bronchial Neoplasms::Carcinoma, Bronchogenic::Carcinoma, Non-Small-Cell Lung [DISEASES] ,neoplasias::neoplasias por localización::neoplasias torácicas::neoplasias del tracto respiratorio::neoplasias pulmonares::neoplasias de los bronquios::carcinoma broncogénico::carcinoma de pulmón de células no pequeñas [ENFERMEDADES] ,Other subheadings::Other subheadings::/drug therapy [Other subheadings] ,digestive system diseases ,Quimioteràpia combinada ,respiratory tract diseases ,Oncology ,binimetinib ,BRAF(V600) mutation ,neoplasms ,Pulmons - Càncer - Tractament ,non-small cell lung cancer ,objective response rate - Abstract
BRAF(V600) oncogenic driver mutations occur in 1-2% of non-small cell lung cancers (NSCLCs) and have been shown to be a clinically relevant target. Preclinical/clinical evidence support the efficacy and safety of BRAF and MEK inhibitor combinations in patients with NSCLC with these mutations. We describe the design of PHAROS, an ongoing, open-label, single-arm, Phase II trial evaluating the BRAF inhibitor encorafenib plus the MEK inhibitor binimetinib in patients with metastatic BRAF(V600) -mutant NSCLC, as first- or second-line treatment. The primary endpoint is objective response rate, based on independent radiologic review (per RECIST v1.1); secondary objectives evaluated additional efficacy endpoints and safety. Results from PHAROS will describe the antitumor activity/safety of encorafenib plus binimetinib in patients with metastatic BRAF(V600)-mutant NSCLC.Lay abstract: Some people with non-small cell lung cancer (NSCLC) have changes in a gene called BRAF (known as 'gene mutations'). One common BRAF mutation is called 'V600'. Combinations of medicines that block proteins encoded by mutant BRAF and another gene called MEK can shrink tumors and slow their progression. We describe the design of PHAROS, a clinical trial investigating encorafenib (mutant BRAF inhibitor) combined with binimetinib (MEK inhibitor) in people with BRAF(V600)-mutant NSCLC that had spread to other parts of the body ('metastatic disease'). People are monitored for side effects and to see if their tumor shrunk. PHAROS includes people treated with encorafenib plus binimetinib as their first treatment for metastatic disease, and people whose cancer progressed after previous anticancer therapy.
- Published
- 2021
31. 516 Peripheral and tumoral immune activity in the expansion part of the first-in-human DuoBody®-PD-L1×4–1BB (GEN1046) trial
- Author
-
Özlem Türeci, Maria Jure-Kunkel, Ignacio Melero, Andrés Cervantes, Tahamtan Ahmadi, Suzana Couto, Nora Pencheva, Alexander Muik, Jose Manuel Trigo Perez, Suresh S. Ramalingam, Brandon W. Higgs, Patricia LoRusso, Muhammad Furqan, Emiliano Calvo, Ugur Sahin, Ulf Forssmann, Victor Moreno, Elena Garralda, Eleni Lagkadinou, Daniel Cho, and Santiago Ponce Aix
- Subjects
Pharmacology ,Oncology ,Cancer Research ,medicine.medical_specialty ,biology ,Combination therapy ,business.industry ,medicine.medical_treatment ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,Institutional review board ,Blockade ,Immune system ,Immunophenotyping ,Informed consent ,Internal medicine ,PD-L1 ,biology.protein ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business ,RC254-282 - Abstract
BackgroundDuoBody-PD-L1×4-1BB (GEN1046) is a class-defining, bispecific immunotherapy designed to induce an antitumor immune response by simultaneous and complementary PD-L1 blockade and conditional 4-1BB stimulation. Encouraging clinical activity and manageable safety were observed during dose escalation in the ongoing phase 1/2a trial in patients with advanced solid tumors (NCT03917381). We report exploratory pharmacodynamic analyses and potential biomarkers of response in an expansion cohort of patients with PD-(L)1–R/R NSCLC.MethodsPatients with metastatic/unresectable NSCLC who had multiple lines of prior systemic therapy, including a checkpoint inhibitor, received flat-dose DuoBody-PD-L1×4-1BB (100 mg) intravenously every 3 weeks. Immunophenotyping of peripheral blood and measurements of soluble immune mediators were evaluated in serial blood samples in cycles 1–2. Tumor PD-L1 and 4-1BB expression and additional immune markers were evaluated by immunohistochemistry in core needle tumor biopsy specimens collected before treatment and at cycle 2.ResultsAs of May 2021, 40 patients with PD-(L)1–R/R NSCLC were enrolled (median age, 63 years). Treatment with DuoBody-PD-L1×4-1BB elicited pharmacodynamic modulation of immune endpoints within the first 2 cycles. Induction of peripheral IFN-y, CXCL9/10, and expansion of peripheral CD8+ effector memory T cells and activated NK cells were observed starting at cycle 1 (>2-fold from baseline) and maintained or increased through cycle 2. Based on 9 paired tumor biopsy samples, increased PD-L1 and 4-1BB expression and cytotoxic CD8+/GZMB+ cell density were detected following treatment. In a subset of patients with clinical response (n=5 confirmed PRs), a trend of greater induction of IFN-y, CXCL9/10, and activated NK cells was observed vs nonresponders. Disease control rates were higher in patients who had progressed on prior anti–PD-1 therapy within 8 months (64% [16/25]) from the first dose of DuoBody-PD-L1×4-1BB. As expected, among patients with evaluable baseline tumors (n=26), most with any degree of tumor reduction (best change, ConclusionsIn patients with NSCLC who progressed on PD-(L)1 therapy, DuoBody-PD-L1×4-1BB elicited pharmacodynamic effects consistent with its proposed mechanism of action. Relationships between disease control and PD-L1 tumoral expression, as well as time from last prior anti–PD-1 therapy, were observed. These findings support that patient selection and/or anti–PD-1 combination therapy may lead to improved clinical efficacy. Further analyses are ongoing and updated results will be presented.AcknowledgementsThe authors thank Hrefna Kristin Johannsdottir, Lei Pang, and Kate Sasser at Genmab A/S and Friederike Gieseke at BioNTech SE for their valuable contributions. This trial was funded by Genmab A/S and BioNTech SE.Trial RegistrationNCT03917381Ethics ApprovalThis trial is undertaken following full approval of the final protocol, amendments, informed consent form, applicable recruiting materials, and subject compensation programs by the Independent Ethics Committee/Institutional Review Board.ConsentWritten informed consent, in accordance with principles that originated in the Declaration of Helsinki 2013, current ICH guidelines including ICH-GCP E6(R2), applicable regulatory requirements, and sponsor policy, was provided by the patients.
- Published
- 2021
32. Pharmacological rescue of tumor intrinsic STING expression and immune response in LKB1-mutant lung cancer via the IAP-JAK regulatory axis
- Author
-
Dacheng Fan, Kavita M. Dhodapkar, Haian Fu, Qiankun Niu, Suresh S. Ramalingam, Madhav V. Dhodapkar, Danielle Cicka, Jennifer W Carlisle, Wei Zhou, Rui Jin, Changfa Shu, Deon B. Doxie, Gabriel Sica, Sean P. Doyle, Andrey A. Ivanov, Alafate Wahafu, Xiulei Mo, Yuhong Du, Xi Zheng, and Taofeek K. Owonikoko
- Subjects
business.industry ,medicine.medical_treatment ,Cancer ,Immunotherapy ,medicine.disease ,Inhibitor of apoptosis ,Phenotype ,Sting ,Immune system ,medicine ,Cancer research ,Cytotoxic T cell ,business ,Lung cancer - Abstract
SummaryHarnessing the power of the immune system to treat cancer has become a core clinical approach. However, rewiring of intrinsic circuitry enables tumor cells to escape immune attacks, leading to therapeutic failure. Pharmacological strategies to reverse tumor genotype-dictated therapeutic resistance are urgently needed to advance precision immunotherapy. Here, we identify antagonists of Inhibitor of Apoptosis Protein (IAP) as potent sensitizers that restore immune-dependent killing of LKB1-mutant lung cancer cells. Mechanistic studies reveal an LKB1-IAP-JAK trimolecular complex that bridges the LKB1-mutant genotype with IAP-dependency and a STING-deficiency-mediated immune resistance phenotype. Ultimately, inhibition of IAP re-establishes JAK-regulated STING expression and DNA sensing pathway as well as enhanced cytotoxic immune cell infiltration and selective immune-dependent anti-tumor activity in an LKB1-mutant immune-competent mouse model. Thus, IAP-JAK-modulatory strategies, like IAP inhibitors, offer promising immunotherapy adjuvants to re-establish the responsiveness of “immunologically-cold” LKB1-mutant tumors to immune checkpoint inhibitors or STING-directed therapies.
- Published
- 2021
33. Correction to: Therapeutic efficacy of the novel SHP2 degrader SHP2-D26, alone or in combination, against lung cancer is associated with modulation of p70S6K/S6, Bim and Mcl-1
- Author
-
Yunfu Deng, Guangzhi Ma, Karin A. Vallega, Dongsheng Wang, Mingliang Wang, Changwei Wang, Shaomeng Wang, Suresh S. Ramalingam, and Shi-Yong Sun
- Subjects
Cancer Research ,Molecular Medicine ,Molecular Biology - Published
- 2022
34. Veliparib in Combination With Platinum-Based Chemotherapy for First-Line Treatment of Advanced Squamous Cell Lung Cancer: A Randomized, Multicenter Phase III Study
- Author
-
Suresh S. Ramalingam, Lei He, Mária Szilasi, Martin Dunbar, Bruce A. Bach, Ramaswamy Govindan, Julien Mazieres, Silvia Novello, R. Bernabé, Xin Huang, Everett E. Vokes, Peter Ansell, Salih Zeki Guclu, Dmitry Bentsion, Philip Clingan, Zanete Zvirbule, Jair Bar, Vasudha Sehgal, Konstantinos N. Syrigos, Jaimee Glasgow, and Lauren Averett Byers
- Subjects
Adult ,Male ,Cancer Research ,Aged ,Aged, 80 and over ,Antineoplastic Combined Chemotherapy Protocols ,Benzimidazoles ,Biomarkers, Tumor ,Carboplatin ,Carcinoma, Non-Small-Cell Lung ,Carcinoma, Squamous Cell ,Clinical Decision-Making ,Female ,Gene Expression Profiling ,Humans ,Lung Neoplasms ,Middle Aged ,Neoplasm Staging ,Paclitaxel ,Patient Selection ,Progression-Free Survival ,Smokers ,Time Factors ,Transcriptome ,Veliparib ,DNA damage ,medicine.medical_treatment ,Squamous cell lung cancer ,chemistry.chemical_compound ,80 and over ,Medicine ,Lung cancer ,Non-Small-Cell Lung ,Polymerase ,Chemotherapy ,Tumor ,biology ,business.industry ,Carcinoma ,medicine.disease ,First line treatment ,Oncology ,chemistry ,Squamous Cell ,biology.protein ,Cancer research ,business ,Biomarkers - Abstract
PURPOSE Squamous non–small-cell lung cancer (sqNSCLC) is genetically complex with evidence of DNA damage. This phase III study investigated the efficacy and safety of poly (ADP-ribose) polymerase inhibitor veliparib in combination with conventional chemotherapy for advanced sqNSCLC ( NCT02106546 ). PATIENTS AND METHODS Patients age ≥ 18 years with untreated, advanced sqNSCLC were randomly assigned 1:1 to carboplatin and paclitaxel with veliparib 120 mg twice daily (twice a day) or placebo twice a day for up to six cycles. The primary end point was overall survival (OS) in the veliparib arm versus the control arm in current smokers, based on phase II findings. Archival tumor samples were provided for biomarker analysis using a 52-gene expression histology classifier (LP52). RESULTS Overall, 970 patients were randomly assigned to carboplatin and paclitaxel plus either veliparib (n = 486) or placebo (n = 484); 57% were current smokers. There was no significant OS benefit with veliparib in current smokers, with median OS 11.9 versus 11.1 months (hazard ratio [HR], 0.905; 95% CI, 0.744 to 1.101; P = .266). In the overall population, OS favored veliparib; median OS was 12.2 versus 11.2 months (HR, 0.853; 95% CI, 0.747 to 0.974), with no difference in progression-free survival (median 5.6 months per arm). In patients with biomarker-evaluable tumor samples (n = 360), OS favored veliparib in the LP52-positive population (median 14.0 v 9.6 months; HR, 0.66; 95% CI, 0.49 to 0.89), but favored placebo in the LP52-negative population (median 11.0 v 14.4 months; HR, 1.33; 95% CI, 0.95 to 1.86). No new safety signals were observed in the experimental arm. CONCLUSION In current smokers with advanced sqNSCLC, there was no therapeutic benefit of adding veliparib to first-line chemotherapy. The LP52 signature may identify a subgroup of patients likely to derive benefit from veliparib with chemotherapy.
- Published
- 2021
35. Overcoming acquired resistance to third-generation EGFR inhibitors by targeting activation of intrinsic apoptotic pathway through Mcl-1 inhibition, Bax activation, or both
- Author
-
Guangzhi Ma, Yunfu Deng, Luxi Qian, Karin A. Vallega, Guojing Zhang, Xingming Deng, Taofeek K. Owonikoko, Suresh S. Ramalingam, Douglas D. Fang, Yifan Zhai, and Shi-Yong Sun
- Subjects
Cancer Research ,Aniline Compounds ,Lung Neoplasms ,Apoptosis ,Article ,ErbB Receptors ,Drug Resistance, Neoplasm ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Mutation ,Genetics ,Humans ,Molecular Biology ,Protein Kinase Inhibitors ,bcl-2-Associated X Protein - Abstract
Treatment of EGFR-mutant non-small cell lung cancer (NSCLC) with mutation-selective third generation EGFR-tyrosine kinase inhibitors (EGFR-TKIs) such as osimertinib has achieved remarkable success in the clinic. However, the immediate challenge is the emergence of acquired resistance, limiting the long-term remission of patients. This study suggests a novel strategy to overcome acquired resistance to osimertinib and other third generation EGFR-TKIs through directly targeting the intrinsic apoptotic pathway. We found that osimertinib, when combined with Mcl-1 inhibition or Bax activation, synergistically decreased the survival of different osimertinib-resistant cell lines, enhanced the induction of intrinsic apoptosis, and inhibited the growth of osimertinib-resistant tumor in vivo. Interestingly, the triple- combination of osimertinib with Mcl-1 inhibition and Bax activation exhibited the most potent activity in decreasing the survival and inducing apoptosis of osimertinib-resistant cells and in suppressing the growth of osimertinib-resistant tumors. These effects were associated with increased activation of the intrinsic apoptotic pathway evidenced by augmented mitochondrial cytochrome C and Smac release. Hence, this study convincingly demonstrates a novel strategy for overcoming acquired resistance to osimertinib and other 3rd generation EGFR-TKIs by targeting activation of the intrinsic apoptotic pathway through Mcl-1 inhibition, Bax activation or both, warranting further clinical validation of this strategy.
- Published
- 2021
36. The CD8+ T cell landscape of human brain metastases
- Author
-
Lisa J. Sudmeier, Matthew Schniederjan, Kimberly B Hoang, Rafi Ahmed, Andreas Wieland, Jeffrey J. Olson, Stewart G. Neill, William H. Hudson, Suresh S. Ramalingam, and Edjah K. Nduom
- Subjects
medicine.anatomical_structure ,Antigen ,T cell ,T-cell receptor ,Cancer research ,medicine ,Cytotoxic T cell ,Biology ,Interleukin-7 receptor ,medicine.disease ,CD8 ,Immune checkpoint ,Brain metastasis - Abstract
Despite improved outcomes with checkpoint blockade immunotherapy, patients with brain metastases have the worst prognosis among patients with metastatic cancer. Immune checkpoint blockade agents target inhibitory receptors, such as PD-1, on exhausted CD8+ T cells to restore their anti-cancer function. Many patients, however, either do not respond or progress after an initial response to immune checkpoint blockade, and distant intracranial failure is common despite excellent options for local treatment of brain metastasis. To develop more effective therapeutic strategies for the treatment of brain metastases, an understanding of the phenotype of brain metastasis-infiltrating CD8+ T cells is essential. Here we performed a detailed characterization of the CD8+ T cells contained in brain metastases. Brain metastases were densely infiltrated by CD8+ T cells; blood contamination of tumor samples was rare. Compared to patient-matched circulating cells, brain metastasis-infiltrating CD8+ T cells had a distinct phenotype characterized by more frequent expression of PD-1, with subpopulations defined by expression of additional co-inhibitory molecules and the residence marker CD69. Single cell RNA-sequencing identified four phenotypic subpopulations within brain metastasis-infiltrating PD-1+ CD8+ T cells. Two of these populations — a terminally-differentiated and a dividing population — were characterized by high expression of co-inhibitory molecules and lacked expression of progenitor markers such as TCF-1. There was significant T cell receptor (TCR) overlap between the terminally-differentiated and dividing populations, suggesting that the dividing cells give rise to the terminally-differentiated cells. There was minimal TCR overlap between these two populations and other brain metastasis-infiltrating PD-1+ CD8+ T cells. T cell clones from brain metastasis-infiltrating CD8+ T cells were rare in circulation, particularly clones from the terminally-differentiated and dividing populations. We systematically identified bystander CD8+ T cells specific for microbial antigens; these cells infiltrated brain metastases and expressed genes shared with exhausted progenitor CD8+ T cells, such as TCF7 and IL7R. We performed spatial transcriptomics on brain metastases and used a novel method to obtain TCR sequences from spatial transcriptomics data. These data revealed distinct niches within the TME defined by their gene expression patterns and cytokine profiles. Terminally-differentiated CD8+ T cells preferentially occupied niches within the tumor parenchyma. Together, our results show that antigen-specificity restricts the spatial localization, phenotypic states, and differentiation pathways available to CD8+ T cells within the brain metastasis TME.
- Published
- 2021
37. Targeting c-Myc to overcome acquired resistance of EGFR mutant NSCLC cells to the third generation EGFR tyrosine kinase inhibitor, osimertinib
- Author
-
Jiajia Gu, Suresh S. Ramalingam, Yong He, Qiming Wang, Lei Zhu, Guojing Zhang, Zhen Chen, Taofeek K. Owonikoko, Hongjing Zang, Shi-Yong Sun, Songqing Fan, and Kevin D.-Y. Sun
- Subjects
Cancer Research ,Proteasome Endopeptidase Complex ,Mutant ,Apoptosis ,Protein degradation ,medicine.disease_cause ,Article ,Proto-Oncogene Proteins c-myc ,T790M ,Cell Line, Tumor ,Medicine ,Animals ,Humans ,Osimertinib ,Protein Kinase Inhibitors ,EGFR inhibitors ,Mutation ,Gene knockdown ,Acrylamides ,Aniline Compounds ,business.industry ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,ErbB Receptors ,Disease Models, Animal ,Oncology ,Mechanism of action ,Drug Resistance, Neoplasm ,Cancer research ,medicine.symptom ,business ,Biomarkers - Abstract
Osimertinib (AZD9291 or TAGRISSO) is a promising and approved third-generation EGFR tyrosine kinase inhibitor (TKI) for treating patients with advanced non–small cell lung cancer (NSCLC) harboring EGFR-activating mutations or the resistant T790M mutation. However, the inevitable emergence of acquired resistance limits its long-term efficacy. A fuller understanding of the mechanism of action of osimertinib and its linkage to acquired resistance will enable the development of more efficacious therapeutic strategies. Consequently, we have identified a novel connection between osimertinib or other EGFR-TKIs and c-Myc. Osimertinib rapidly and sustainably decreased c-Myc levels primarily via enhancing protein degradation in EGFR-mutant (EGFRm) NSCLC cell lines and xenograft tumors. c-Myc levels were substantially elevated in different EGFRm NSCLC cell lines with acquired resistance to osimertinib in comparison with their corresponding parental cell lines and could not be reduced any further by osimertinib. Consistently, c-Myc levels were elevated in the majority of EGFRm NSCLC tissues relapsed from EGFR-TKI treatment compared with their corresponding untreated baseline c-Myc levels. Suppression of c-Myc through knockdown or pharmacologic targeting with BET inhibitors restored the response of resistant cell lines to osimertinib. These findings indicate that c-Myc modulation mediates the therapeutic efficacy of osimertinib and the development of osimertinib acquired resistance. Furthermore, they establish c-Myc as a potential therapeutic target and warrant clinical testing of BET inhibition as a potential strategy to overcome acquired resistance to osimertinib or other EGFR inhibitors. Significance: This study demonstrates a critical role of c-Myc modulation in mediating therapeutic efficacy of osimertinib including osimertinib acquired resistance and suggests targeting c-Myc as a potential strategy to overcome osimertinib acquired resistance.
- Published
- 2021
38. JASPER: Phase 2 trial of first-line niraparib plus pembrolizumab in patients with advanced non-small cell lung cancer
- Author
-
Grace K. Dy, Iryna Teslenko, Sharon Lu, Eddie Thara, Yongqiang Tang, Afshin Dowlati, David R. Spigel, Nithya Iyer Singh, Suresh S. Ramalingam, Thomas E. Stinchcombe, Basir Haque, Nicholas Iannotti, and Mark M. Awad
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Indazoles ,Lung Neoplasms ,business.industry ,Pembrolizumab ,Poly(ADP-ribose) Polymerase Inhibitors ,medicine.disease ,Antibodies, Monoclonal, Humanized ,Clinical trial ,Piperidines ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Cohort ,PARP inhibitor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,business ,Lung cancer ,Adverse effect ,Tumor marker - Abstract
Background Poly(ADP-ribose) polymerase (PARP) inhibitors may synergize with programmed cell death receptor-1 (PD-1) inhibitors to enhance adaptive and innate antitumor immune responses. In the phase 2 JASPER study (NCT04475939), the PARP inhibitor niraparib was evaluated in combination with the PD-1 inhibitor pembrolizumab in patients with metastatic and/or locally advanced non-small cell lung cancer (NSCLC). Methods Patients whose tumors had programmed cell death ligand 1 (PD-L1) tumor proportion scores (TPS) ≥50% (cohort 1) or 1%-49% (cohort 2) received first-line niraparib (200 mg once daily) plus pembrolizumab (200 mg every 3 weeks). The primary end point was investigator-assessed objective response rate (ORR). Secondary end points included duration of response (DoR), progression-free survival (PFS), overall survival (OS), safety, and pharmacokinetics. Results Thirty-eight patients were enrolled in cohorts 1 and 2. In cohort 1, ORR (95% confidence interval [CI]) was 56.3% (9 of 16 patients; 29.9%-80.2%); 2 of 16 patients had complete responses and 7 of 16 had partial responses (PRs). In cohort 2, ORR was 20.0% (5.7%-43.7%) with 4 of 20 PRs. In cohorts 1 and 2, the median DoR was 19.7 months (95% CI, 4.2 months to not estimable [NE]) and 9.4 months (95% CI, 4.2 months to NE), the median PFS was 8.4 months (95% CI, 3.9-22.1 months) and 4.2 months (95% CI, 2.0-6.2 months), and the median OS was NE (95% CI, 6.0 months to NE) and 7.7 months (95% CI, 4.0-12.5 months), respectively. Grade ≥3 treatment-emergent adverse events occurred in 88.2% and 85.7% of patients in cohorts 1 and 2, respectively. Safety was consistent with known profiles of single-agent niraparib and pembrolizumab. Conclusions Niraparib plus pembrolizumab showed clinical activity in patients with advanced and/or metastatic NSCLC. Lay summary The JASPER clinical trial studied a new combination treatment for advanced or metastatic non-small cell lung cancer (NSCLC). Pembrolizumab, a drug approved for NSCLC, was given with niraparib. Previous research showed that these 2 drugs together might work better than either drug alone. This study found that more than half of patients with high levels of a tumor marker responded to the combination, and one-fifth of patients with lower levels of the marker responded. The types of side effects from the combination were similar to side effects from both drugs alone. These results support more research on this combination.
- Published
- 2021
39. Nivolumab Plus Ipilimumab vs Nivolumab for Previously Treated Patients With Stage IV Squamous Cell Lung Cancer
- Author
-
Lyudmila Bazhenova, Scott N. Gettinger, Fred R. Hirsch, Mary W. Redman, Thomas E. Stinchcombe, Susan S. Tavernier, Hui Yu, Karen Kelly, Lawrence H. Schwartz, Jeffrey D. Bradley, Roy S. Herbst, Philip C. Mack, Katherine Minichiello, Louise Highleyman, Vassiliki A. Papadimitrakopoulou, David R. Gandara, Natasha B. Leighl, Suresh S. Ramalingam, and Joseph M. Unger
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Ipilimumab ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Online First ,Lung cancer ,Lung ,Original Investigation ,business.industry ,Research ,Epithelial Cells ,medicine.disease ,Interim analysis ,Chemotherapy regimen ,Nivolumab ,Docetaxel ,Response Evaluation Criteria in Solid Tumors ,Carcinoma, Squamous Cell ,business ,Comments ,medicine.drug - Abstract
This phase 3 randomized clinical trial investigates if the addition of ipilimumab to nivolumab improves the survival outcome of patients with chemotherapy-pretreated immunotherapy-naive squamous cell lung cancer., Key Points Question Does the addition of ipilimumab to nivolumab improve survival in patients with advanced chemotherapy-pretreated immunotherapy-naive squamous cell lung cancer? Findings In this randomized clinical trial of 252 patients, the addition of ipilimumab to nivolumab did not lead to improved survival in patients with advanced chemotherapy-pretreated squamous cell carcinoma. Meaning Combination therapy with nivolumab and ipilimumab is currently only indicated as first-line therapy in patients with advanced non–small cell lung cancer., Importance Nivolumab plus ipilimumab is superior to platinum-based chemotherapy in treatment-naive advanced non–small cell lung cancer (NSCLC). Nivolumab is superior to docetaxel in advanced pretreated NSCLC. Objective To determine whether the addition of ipilimumab to nivolumab improves survival in patients with advanced, pretreated, immunotherapy-naive squamous (Sq) NSCLC. Design, Setting, and Participants The Lung Cancer Master Protocol (Lung-MAP) S1400I phase 3, open-label randomized clinical trial was conducted from December 18, 2015, to April 23, 2018, randomizing patients in a 1:1 ratio to nivolumab alone or combined with ipilimumab. The median follow-up in surviving patients was 29.5 months. The trial was conducted through the National Clinical Trials Network and included patients with advanced immunotherapy-naive SqNSCLC and a Zubrod score of 0 (asymptomatic) to 1 (symptomatic but completely ambulatory) with disease progression after standard platinum-based chemotherapy. Randomization was stratified by sex and number of prior therapies (1 vs 2 or more). Data were analyzed from May 3, 2018, to February 1, 2021. Interventions Nivolumab, 3 mg/kg intravenously every 2 weeks, with or without ipilimumab, 1 mg/kg intravenously every 6 weeks, until disease progression or intolerable toxic effects. Main Outcomes and Measures The primary end point was overall survival (OS). Secondary end points included investigator-assessed progression-free survival (IA-PFS) and response per Response Evaluation Criteria in Solid Tumors (RECIST) guidelines, version 1.1. Results Of 275 enrolled patients, 252 (mean age, 67.5 years [range 41.8-90.3 years]; 169 men [67%]; 206 White patients [82%]) were deemed eligible (125 randomized to nivolumab/ipilimumab and 127 to nivolumab). The study was closed for futility at a planned interim analysis. Overall survival was not significantly different between the groups (hazard ratio [HR], 0.87; 95% CI, 0.66-1.16; P = .34). Median survival was 10 months (95% CI, 8.0-14.4 months) in the nivolumab/ipilimumab group and 11 months (95% CI, 8.6-13.7 months) in the nivolumab group. The IA-PFS HR was 0.80 (95% CI, 0.61-1.03; P = .09); median IA-PFS was 3.8 months (95% CI, 2.7-4.4 months) in the nivolumab/ipilimumab group and 2.9 months (95% CI, 1.8-4.0 months) in the nivolumab alone group. Response rates were 18% (95% CI, 12%-25%) with nivolumab/ipilimumab and 17% (95% CI, 10%-23%) with nivolumab. Median response duration was 28.4 months (95% CI, 4.9 months to not reached) with nivolumab/ipilimumab and 9.7 months with nivolumab (95% CI, 4.2-23.1 months). Grade 3 or higher treatment-related adverse events occurred in 49 of 124 patients (39.5%) who received nivolumab/ipilimumab and in 41 of 123 (33.3%) who received nivolumab alone. Toxic effects led to discontinuation in 31 of 124 patients (25%) on nivolumab/ipilimumab and in 19 of 123 (15%) on nivolumab. Conclusions and Relevance In this phase 3 randomized clinical trial, ipilimumab added to nivolumab did not improve outcomes in patients with advanced, pretreated, immune checkpoint inhibitor–naive SqNSCLC. Trial Registration ClinicalTrials.gov Identifier: NCT02785952
- Published
- 2021
40. Epidermal growth factor receptor mutation analysis in tissue and plasma from the AURA3 trial: Osimertinib versus platinum‐pemetrexed for T790M mutation‐positive advanced non–small cell lung cancer
- Author
-
Sabina Patel, Chun-Ming Tsai, X. Huang, Suzanne Jenkins, Makoto Maemondo, Kenneth S. Thress, Aleksandra Markovets, G. Laus, Yi-Long Wu, Yong He, Sang We Kim, Janessa Laskin, Toyoaki Hida, Terufumi Kato, Angelo Delmonte, Suresh S. Ramalingam, Tony Mok, Te Chun Hsia, Ji Youn Han, Vassiliki A. Papadimitrakopoulou, and Myung-Ju Ahn
- Subjects
Male ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,DNA Mutational Analysis ,non-small cell lung cancer (NSCLC) ,Pemetrexed ,Carboplatin ,Circulating Tumor DNA ,03 medical and health sciences ,T790M ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Biopsy ,Humans ,Medicine ,Osimertinib ,Digital polymerase chain reaction ,030212 general & internal medicine ,Epidermal growth factor receptor ,Lung cancer ,Lung ,Retrospective Studies ,Acrylamides ,Aniline Compounds ,medicine.diagnostic_test ,biology ,business.industry ,Middle Aged ,medicine.disease ,Progression-Free Survival ,Tumor Burden ,respiratory tract diseases ,ErbB Receptors ,030220 oncology & carcinogenesis ,Mutation ,biology.protein ,Female ,Cisplatin ,business ,medicine.drug - Abstract
BACKGROUND This study assesses different technologies for detecting epidermal growth factor receptor (EGFR) mutations from circulating tumor DNA in patients with EGFR T790M-positive advanced non-small cell lung cancer (NSCLC) from the AURA3 study (NCT02151981), and it evaluates clinical responses to osimertinib and platinum-pemetrexed according to the plasma T790M status. METHODS Tumor tissue biopsy samples were tested for T790M during screening with the cobas EGFR Mutation Test (cobas tissue). Plasma samples were collected at screening and at the baseline and were retrospectively analyzed for EGFR mutations with the cobas EGFR Mutation Test v2 (cobas plasma), droplet digital polymerase chain reaction (ddPCR; Biodesix), and next-generation sequencing (NGS; Guardant360, Guardant Health). RESULTS With cobas tissue test results as a reference, the plasma T790M positive percent agreement (PPA) was 51% (110 of 215 samples) by cobas plasma, 58% (110 of 189) by ddPCR, and 66% (136 of 207) by NGS. Plasma T790M detection was associated with a larger median baseline tumor size (56 mm for T790M-positive vs 39 mm for T790M-negative; P
- Published
- 2019
41. Tissue and Plasma EGFR Mutation Analysis in the FLAURA Trial: Osimertinib versus Comparator EGFR Tyrosine Kinase Inhibitor as First-Line Treatment in Patients with EGFR-Mutated Advanced Non–Small Cell Lung Cancer
- Author
-
Fumio Imamura, Suresh S. Ramalingam, Xiaocheng Li-Sucholeiki, Eun Kyung Cho, X. Huang, Jhanelle E. Gray, Manuel Cobo, Wu Chou Su, Kazuo Kasahara, Suzanne Jenkins, Virote Sriuranpong, Isamu Okamoto, Guili Zhang, Yuri Rukazenkov, Simon Dearden, Ying Cheng, Johan Vansteenkiste, Yong Kek Pang, M. Saggese, Naoyuki Nogami, Brian Lentrichia, and Jong Seok Lee
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Randomization ,Clinical Decision-Making ,Antineoplastic Agents ,Article ,03 medical and health sciences ,0302 clinical medicine ,Carcinoma, Non-Small-Cell Lung ,Internal medicine ,medicine ,Carcinoma ,Humans ,Osimertinib ,Neoplasm Metastasis ,Lung cancer ,Protein Kinase Inhibitors ,Alleles ,Neoplasm Staging ,Randomized Controlled Trials as Topic ,Acrylamides ,Aniline Compounds ,business.industry ,Disease Management ,medicine.disease ,Confidence interval ,ErbB Receptors ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,Amino Acid Substitution ,Clinical Trials, Phase III as Topic ,Egfr mutation ,030220 oncology & carcinogenesis ,Mutation ,Female ,Non small cell ,business - Abstract
Purpose: To assess the utility of the cobas EGFR Mutation Test, with tissue and plasma, for first-line osimertinib therapy for patients with EGFR-mutated (EGFRm; Ex19del and/or L858R) advanced or metastatic non–small cell lung cancer (NSCLC) from the FLAURA study (NCT02296125). Experimental Design: Tumor tissue EGFRm status was determined at screening using the central cobas tissue test or a local tissue test. Baseline circulating tumor (ct)DNA EGFRm status was retrospectively determined with the central cobas plasma test. Results: Of 994 patients screened, 556 were randomized (289 and 267 with central and local EGFR test results, respectively) and 438 failed screening. Of those randomized from local EGFR test results, 217 patients had available central test results; 211/217 (97%) were retrospectively confirmed EGFRm positive by central cobas tissue test. Using reference central cobas tissue test results, positive percent agreements with cobas plasma test results for Ex19del and L858R detection were 79% [95% confidence interval (CI), 74–84] and 68% (95% CI, 61–75), respectively. Progression-free survival (PFS) superiority with osimertinib over comparator EGFR-TKI remained consistent irrespective of randomization route (central/local EGFRm-positive tissue test). In both treatment arms, PFS was prolonged in plasma ctDNA EGFRm-negative (23.5 and 15.0 months) versus -positive patients (15.2 and 9.7 months). Conclusions: Our results support utility of cobas tissue and plasma testing to aid selection of patients with EGFRm advanced NSCLC for first-line osimertinib treatment. Lack of EGFRm detection in plasma was associated with prolonged PFS versus patients plasma EGFRm positive, potentially due to patients having lower tumor burden.
- Published
- 2019
42. Adiposity may predict survival in patients with advanced stage cancer treated with immunotherapy in phase 1 clinical trials
- Author
-
Milton Williams, Viraj A. Master, Olatunji B. Alese, David H. Lawson, Christina Wu, Ragini R. Kudchadkar, Hannah Collins, Yuan Liu, Amir Ishaq Khan, Dylan J. Martini, Conor E. Steuer, Haydn T. Kissick, Mehmet Akce, Bradley C. Carthon, Bassel F. El-Rayes, R. Donald Harvey, Colleen Lewis, Julie M. Shabto, Suresh S. Ramalingam, Taofeek K. Owonikoko, Meredith R Kline, Mehmet Asim Bilen, Rathi N. Pillai, and Walid L. Shaib
- Subjects
Cancer Research ,medicine.medical_specialty ,business.industry ,Proportional hazards model ,Concordance ,Hazard ratio ,Phases of clinical research ,Cancer ,Recursive partitioning ,medicine.disease ,Gastroenterology ,Obesity ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,030212 general & internal medicine ,business ,Body mass index - Abstract
BACKGROUND Body mass index (BMI) is used to define obesity, but it is an imperfect measure of body composition. In the current study, the authors explored the association between types of fat and survival in patients treated with immunotherapy. METHODS A retrospective analysis of 90 patients who were treated with immunotherapy on phase 1 clinical trials at the Winship Cancer Institute in Atlanta, Georgia, from 2009 through 2017 was performed. Overall survival (OS) and progression-free survival (PFS) were used to measure clinical outcomes. Baseline BMI and radiographic images at the middle of the third lumbar vertebrae were obtained. Fat densities were calculated and converted to indices (subcutaneous fat index [SFI], intermuscular fat index [IFI], and visceral fat index [VFI]) after dividing by height in meters squared. Risk groups were created using recursive partitioning and the regression trees method for SFI and IFI, which were selected by stepwise variable selection among all fat-related variables. The Cox proportional hazards model and Kaplan-Meier method were used for the association with OS and PFS. RESULTS The majority of patients (59%) were male and diagnosed with melanoma (33%) or gastrointestinal cancers (22%). The median BMI was 27.4 kg/m2 , the median SFI was 62.78, the median IFI was 4.06, and the median VFI was 40.53. Low-risk patients (those with an SFI ≥73) had a significantly longer OS (hazard ratio, 0.20; 95% CI, 0.09-0.46 [P
- Published
- 2019
43. Five-Year Overall Survival for Patients With Advanced Non‒Small-Cell Lung Cancer Treated With Pembrolizumab: Results From the Phase I KEYNOTE-001 Study
- Author
-
Matthew D. Hellmann, Myung-Ju Ahn, Ani Sarkis Balmanoukian, Edward B. Garon, Jonathan W. Goldman, Enric Carcereny, Joseph Paul Eder, Debra Kush, Erin Jensen, Naiyer A. Rizvi, Leora Horn, Suresh S. Ramalingam, Charu Aggarwal, Natasha B. Leighl, Rina Hui, Matthew A. Gubens, Enriqueta Felip, Amita Patnaik, and Cathie Scalzo
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Lung Cancer ,Pembrolizumab ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Monoclonal ,Carcinoma ,medicine ,Overall survival ,Non small cell ,RAPID COMMUNICATION ,business ,Lung cancer ,Survival rate - Abstract
PURPOSE Pembrolizumab monotherapy has demonstrated durable antitumor activity in advanced programmed death ligand 1 (PD-L1)–expressing non‒small-cell lung cancer (NSCLC). We report 5-year outcomes from the phase Ib KEYNOTE-001 study. These data provide the longest efficacy and safety follow-up for patients with NSCLC treated with pembrolizumab monotherapy. PATIENTS AND METHODS Eligible patients had confirmed locally advanced/metastatic NSCLC and provided a contemporaneous tumor sample for PD-L1 evaluation by immunohistochemistry using the 22C3 antibody. Patients received intravenous pembrolizumab 2 mg/kg every 3 weeks or 10 mg/kg every 2 or 3 weeks. Investigators assessed response per immune-related response criteria. The primary efficacy end point was objective response rate. Overall survival (OS) and duration of response were secondary end points. RESULTS We enrolled 101 treatment-naive and 449 previously treated patients. Median follow-up was 60.6 months (range, 51.8 to 77.9 months). At data cutoff—November 5, 2018—450 patients (82%) had died. Median OS was 22.3 months (95% CI, 17.1 to 32.3 months) in treatment-naive patients and 10.5 months (95% CI, 8.6 to 13.2 months) in previously treated patients. Estimated 5-year OS was 23.2% for treatment-naive patients and 15.5% for previously treated patients. In patients with a PD-L1 tumor proportion score of 50% or greater, 5-year OS was 29.6% and 25.0% in treatment-naive and previously treated patients, respectively. Compared with analysis at 3 years, only three new-onset treatment-related grade 3 adverse events occurred (hypertension, glucose intolerance, and hypersensitivity reaction, all resolved). No late-onset grade 4 or 5 treatment-related adverse events occurred. CONCLUSION Pembrolizumab monotherapy provided durable antitumor activity and high 5-year OS rates in patients with treatment-naive or previously treated advanced NSCLC. Of note, the 5-year OS rate exceeded 25% among patients with a PD-L1 tumor proportion score of 50% or greater. Pembrolizumab had a tolerable long-term safety profile with little evidence of late-onset or new toxicity.
- Published
- 2019
44. DNA-PKc deficiency drives pre-malignant transformation by reducing DNA repair capacity in concert with reprogramming the epigenome in human bronchial epithelial cells
- Author
-
Christopher Dagucon, Shuguang Leng, Suresh S. Ramalingam, Maria A. Picchi, Steven A. Belinsky, Ivo Teneng, and Carmen S. Tellez
- Subjects
DNA Repair ,DNA repair ,DNA damage ,Bronchi ,DNA-Activated Protein Kinase ,Haploinsufficiency ,Respiratory Mucosa ,Biology ,Biochemistry ,Article ,Epigenesis, Genetic ,Bleomycin ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Humans ,Gene silencing ,Epigenetics ,Promoter Regions, Genetic ,Molecular Biology ,Gene ,030304 developmental biology ,0303 health sciences ,Gene knockdown ,Epithelial Cells ,Cell Biology ,Epigenome ,DNA Methylation ,Cell Transformation, Neoplastic ,030220 oncology & carcinogenesis ,Cancer research ,Reprogramming - Abstract
The expression of DNA-dependent protein kinase catalytic subunit (DNA-PKc) is highly variable in smokers and reduced enzyme activity has been associated with risk for lung cancer. An in vitro model of lung pre-malignancy was used to evaluate the role of double-strand break DNA repair capacity in transformation of hTERT/CDK4 immortalized human bronchial epithelial cells (HBECs) and reprograming of the epigenome. Here we show that knockdown of DNA-PKc to levels simulating haploinsufficiency dramatically reduced DNA repair capacity following challenge with bleomycin and significantly increased transformation efficiency of HBEC lines exposed weekly for 12 weeks to this radiomimetic. Transformed HBEC lines with wild type or knockdown of DNA-PKc showed altered expression of more than 1,000 genes linked to major cell regulatory pathways involved in lung cancer. While lung cancer driver mutations were not detected in transformed clones, more than 300 genes that showed reduced expression associated with promoter methylation in transformed clones or predictive for methylation in malignant tumors were identified. These studies support reduced DNA repair capacity as a key factor in the initiation and clonal expansion of pre-neoplastic cells and double-strand break DNA damage as causal for epigenetic mediated silencing of many lung cancer-associated genes. The fact that DNA damage, repair, and epigenetic silencing of genes are causal for many other cancers that include colon and prostate extends the generalizability and impact of these findings.
- Published
- 2019
45. Small Molecule KRAS Agonist for Mutant KRAS Cancer Therapy
- Author
-
Andrew T. Magis, Xingming Deng, Wei Zhou, Dongkyoo Park, Jun Zhang, Walter J. Curran, Suresh S. Ramalingam, Ke Xu, and Gabriel Sica
- Subjects
0301 basic medicine ,Agonist ,Cancer Research ,Programmed cell death ,endocrine system diseases ,medicine.drug_class ,Mutant ,Apoptosis ,Treatment of lung cancer ,Biology ,medicine.disease_cause ,NSCLC ,lcsh:RC254-282 ,03 medical and health sciences ,0302 clinical medicine ,medicine ,KRAS ,Autophagy ,Lung cancer ,neoplasms ,Cell growth ,Research ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,digestive system diseases ,3. Good health ,respiratory tract diseases ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Therapy - Abstract
Background Lung cancer patients with KRAS mutation(s) have a poor prognosis due in part to the development of resistance to currently available therapeutic interventions. Development of a new class of anticancer agents that directly targets KRAS may provide a more attractive option for the treatment of KRAS-mutant lung cancer. Results Here we identified a small molecule KRAS agonist, KRA-533, that binds the GTP/GDP-binding pocket of KRAS. In vitro GDP/GTP exchange assay reveals that KRA-533 activates KRAS by preventing the cleavage of GTP into GDP, leading to the accumulation of GTP-KRAS, an active form of KRAS. Treatment of human lung cancer cells with KRA-533 resulted in increased KRAS activity and suppression of cell growth. Lung cancer cell lines with KRAS mutation were relatively more sensitive to KRA-533 than cell lines without KRAS mutation. Mutating one of the hydrogen-bonds among the KRA-533 binding amino acids in KRAS (mutant K117A) resulted in failure of KRAS to bind KRA-533. KRA-533 had no effect on the activity of K117A mutant KRAS, suggesting that KRA-533 binding to K117 is required for KRA-533 to enhance KRAS activity. Intriguingly, KRA-533-mediated KRAS activation not only promoted apoptosis but also autophagic cell death. In mutant KRAS lung cancer xenografts and genetically engineered mutant KRAS-driven lung cancer models, KRA-533 suppressed malignant growth without significant toxicity to normal tissues. Conclusions The development of this KRAS agonist as a new class of anticancer drug offers a potentially effective strategy for the treatment of lung cancer with KRAS mutation and/or mutant KRAS-driven lung cancer. Electronic supplementary material The online version of this article (10.1186/s12943-019-1012-4) contains supplementary material, which is available to authorized users.
- Published
- 2019
46. The Third-Generation EGFR Inhibitor, Osimertinib, Promotes c-FLIP Degradation, Enhancing Apoptosis Including TRAIL-Induced Apoptosis in NSCLC Cells with Activating EGFR Mutations
- Author
-
Guoqing Qian, Hui Ren, Suresh S. Ramalingam, Lei Zhu, Mingwei Chen, Shi-Yong Sun, Shuo Zhang, and Puyu Shi
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,Original article ,Mutant ,Ligand (biochemistry) ,medicine.disease_cause ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Apoptosis ,Flip ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Osimertinib ,Tumor necrosis factor alpha ,EGFR inhibitors - Abstract
The third-generation EGFR inhibitor, osimertinib (AZD9291), selectively and irreversibly inhibits EGFR activating and T790 M mutants while sparing wild-type EGFR. Osimertinib is now an approved drug for non-small cell lung cancer (NSCLC) patients with activating EGFR mutations (first-line) or those who have become resistant to 1st generation EGFR inhibitors through the T790 M mutation (second-line). Unfortunately, all patients eventually relapse and develop resistance to osimertinib. Hence, it is essential to fully understand the biology underlying the development of resistance to osimertinib in order to improve its therapeutic efficacy and overcome resistance. Cellular FLICE-inhibitory protein (c-FLIP) is a truncated form of caspase-8 and functions as a key inhibitor of the extrinsic apoptotic pathway. The current study has demonstrated that osimertinib reduces c-FLIP levels via facilitating its degradation and enhances apoptosis induced by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) primarily in NSCLC with activating EGFR mutations. Moreover, modulation of c-FLIP expression levels, to some degree, also alters the sensitivities of EGFR mutant NSCLC cells to undergo osimertinib-induced apoptosis, suggesting that c-FLIP suppression is an important event contributing to the antitumor activity of osimertinib against EGFR mutant NSCLC.
- Published
- 2019
47. First-Line Nivolumab Plus Ipilimumab in Advanced Non–Small-Cell Lung Cancer (CheckMate 568): Outcomes by Programmed Death Ligand 1 and Tumor Mutational Burden as Biomarkers
- Author
-
Martin Gutierrez, Mark M. Awad, Joseph D. Szustakowski, Suresh S. Ramalingam, Jason Chesney, Justin F. Gainor, Neal Ready, Kim E. Zerba, Pavan S. Reddy, Ian Rabinowitz, Brian Lestini, Mihaela Mates, Xuemei Li, Martin Reck, James Michael Orcutt, George Green, Julie R. Brahmer, David R. Spigel, William J. Geese, Matthew D. Hellmann, Gregory A. Otterson, Kenneth J. O'Byrne, Jacques Jolivet, Wenhua Hu, Luis Paz-Ares, Leora Horn, Han Chang, and Hossein Borghaei
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,non-small cell lung cancer (NSCLC) ,Ipilimumab ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Carcinoma ,Biomarkers, Tumor ,Humans ,Lung cancer ,Aged ,Neoplasm Staging ,Aged, 80 and over ,business.industry ,Lung Cancer ,ORIGINAL REPORTS ,Middle Aged ,medicine.disease ,Clinical trial ,030104 developmental biology ,Treatment Outcome ,Nivolumab ,Docetaxel ,030220 oncology & carcinogenesis ,Mutation ,Female ,Neoplasm Recurrence, Local ,business ,Biomarkers ,medicine.drug - Abstract
PURPOSE CheckMate 568 is an open-label phase II trial that evaluated the efficacy and safety of nivolumab plus low-dose ipilimumab as first-line treatment of advanced/metastatic non–small-cell lung cancer (NSCLC). We assessed the association of efficacy with programmed death ligand 1 (PD-L1) expression and tumor mutational burden (TMB). PATIENTS AND METHODS Two hundred eighty-eight patients with previously untreated, recurrent stage IIIB/IV NSCLC received nivolumab 3 mg/kg every 2 weeks plus ipilimumab 1 mg/kg every 6 weeks. The primary end point was objective response rate (ORR) in patients with 1% or more and less than 1% tumor PD-L1 expression. Efficacy on the basis of TMB (FoundationOne CDx assay) was a secondary end point. RESULTS Of treated patients with tumor available for testing, 252 patients (88%) of 288 were evaluable for PD-L1 expression and 98 patients (82%) of 120 for TMB. ORR was 30% overall and 41% and 15% in patients with 1% or greater and less than 1% tumor PD-L1 expression, respectively. ORR increased with higher TMB, plateauing at 10 or more mutations/megabase (mut/Mb). Regardless of PD-L1 expression, ORRs were higher in patients with TMB of 10 or more mut/Mb (n = 48: PD-L1, ≥ 1%, 48%; PD-L1, < 1%, 47%) versus TMB of fewer than 10 mut/Mb (n = 50: PD-L1, ≥ 1%, 18%; PD-L1, < 1%, 5%), and progression-free survival was longer in patients with TMB of 10 or more mut/Mb versus TMB of fewer than 10 mut/Mb (median, 7.1 v 2.6 months). Grade 3 to 4 treatment-related adverse events occurred in 29% of patients. CONCLUSION Nivolumab plus low-dose ipilimumab was effective and tolerable as a first-line treatment of advanced/metastatic NSCLC. TMB of 10 or more mut/Mb was associated with improved response and prolonged progression-free survival in both tumor PD-L1 expression 1% or greater and less than 1% subgroups and was thus identified as a potentially relevant cutoff in the assessment of TMB as a biomarker for first-line nivolumab plus ipilimumab.
- Published
- 2019
48. Abstract 428: Therapeutic potential of the novel SHP2 degrader, SHP2-D26, alone or in combination, in the treatment of lung cancer
- Author
-
Yunful Deng, Guangzhi Ma, Karin A. Vallega, Dongsheng Wang, Mingliang Wang, Changwei Wang, Shaomeng Wang, Suresh S. Ramalingam, and Shi-Yong Sun
- Subjects
Cancer Research ,Oncology - Abstract
Src homology 2 domain-containing phosphatase 2 (SHP2) functions as a protein tyrosine phosphatase and plays a critical role in the full activation of the Ras-MAPK signaling pathway downstream of cell surface tyrosine receptors (e.g., EGFR), which are frequently amplified or mutationally activated in human cancer. Therefore, SHP2 has emerged as an attractive therapeutic target for human cancers and other human diseases. While some allosteric inhibitors that are being tested in clinical trials, a few SHP2 degraders have been developed recently. However, their activities in human lung cancer have not been tested. The current study focused on evaluating the therapeutic potential of the novel SHP2 degrader, SHP2-D26 (D26), either alone or in combination, against non-small cell lung cancer cell (NSCLC). Most of tested NSCLC cell lines (12/14) were insensitive to the control small molecule SHP2 inhibitor with IC50s of far over 10 μM. However, all these cell lines responded to D26 with IC50s of < 8 μM. A few cell lines (4/14; e.g. HCC827, PC-9, H1792 and H1648) were much more sensitive than others with IC50s of < 3 μM. There was no clear association between basal levels of SHP2 and cell sensitivities to D26. Interestingly, D26 rapidly and potently decreased SHP2 levels in all the tested cell lines in a sustained way regardless cell sensitivities to D26, suggesting that there may be additional factors that impact cell response to D26. We noted that suppression of p70S6/S6, but not ERK1/2, was associated with cell responses to D26, warranting further study in this direction. In the sensitive cell lines, D26 effectively increased Bim levels while decreasing Mcl-1 levels likely through modulating their stability accompanied with induction of apoptosis. These events are all important for D26 to induce apoptosis. When combined with the third generation EGFR, osimertinib (AZD9291), synergistic effects on decreasing the survival of different osimertinib-resistant cell lines were observed. Moreover, the combination enhanced induction of apoptosis in these osimertinib-resistant cells. Although D26 alone exerted moderate effect on inhibiting the growth of NSCLC xenografts, the combination of osimertinib with D26 very effectively inhibited the growth of osimertinib-resistant xenografts, suggesting promising efficacy in overcoming acquired resistance to osimertinib. Our findings thus warrant further evaluation of the potential of D26 in overcoming acquired resistance to osimertinib in clinic. Citation Format: Yunful Deng, Guangzhi Ma, Karin A. Vallega, Dongsheng Wang, Mingliang Wang, Changwei Wang, Shaomeng Wang, Suresh S. Ramalingam, Shi-Yong Sun. Therapeutic potential of the novel SHP2 degrader, SHP2-D26, alone or in combination, in the treatment of lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 428.
- Published
- 2022
49. Abstract CT008: Long-term outcomes with sotorasib in pretreated KRASp.G12C-mutated NSCLC: 2-year analysis of CodeBreaK100
- Author
-
Grace K. Dy, Ramaswamy Govindan, Vamsidhar Velcheti, Gerald S. Falchook, Antoine Italiano, Juergen Wolf, Adrian G. Sacher, Toshiaki Takahashi, Suresh S. Ramalingam, Christophe Dooms, Dong-Wan Kim, Alfredo Addeo, Jayesh Desai, Martin Schuler, Pascale Tomasini, Qui Tran, Simon Jones, Agnes Ang, Abraham Anderson, Antreas A. Hindoyan, David S. Hong, and Bob T. Li
- Subjects
Cancer Research ,Oncology - Abstract
Introduction: Sotorasib, a first-in-class KRASG12C inhibitor, has been FDA-approved for adults with KRAS p.G12C-mutated locally advanced or metastatic NSCLC who received prior systemic therapies based on the Phase 1/2 global, single-arm CodeBreaK100 trial. We will report the longest follow-up for a KRASG12C inhibitor, including 2-year survival, safety, and genomic profiles associated with durable clinical benefit. Methods: Sotorasib was administered orally at 960 mg once daily to patients who progressed on prior therapies and had KRAS p.G12C-mutated locally advanced, metastatic NSCLC. Primary endpoint was objective response rate (ORR) assessed by central review. Key secondary endpoints included duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. In an exploratory analysis, baseline tumor tissue and/or plasma samples were collected and analyzed for genomic alterations. Tumor samples were analyzed for PD-L1 levels to evaluate correlates with prolonged tumor response, defined as patients with PFS ≥ 12 months, in comparison with those with PFS ≤ 3 months. Results: In the first analysis of the combined Phase 1 and Phase 2 study (N=174), the median number of prior lines of therapy was 2.0 (range 1-4+). 90.2% received prior anti-PD1 or anti PD-L1 treatment; 82.8% received both prior platinum-based chemotherapy and anti-PD1/PD-L1. Updated ORR by central review was 40.7% (95% CI: 33.2, 48.4) and median DOR was 12.3 months (7.1, 14.6). Median PFS and OS was 6.3 months (95% CI: 5.3, 8.2) and 12.5 months (10.0, 17.8). One and two-year OS was 50.8% and 30.3%. Sotorasib was well-tolerated in the long-term with mild and manageable toxicities, and no new safety signals emerged. Prolonged tumor response was observed across PD-L1 expression, including in tumors with low PDL-1 expression and STK11 co-mutation that may derive less benefit from immunotherapy. Conclusions: In the longest follow-up of any KRASG12C inhibitor, sotorasib continued to demonstrate a favorable safety profile and durable efficacy, including a 2-year OS observed in 30% of patients. Current analyses continue to support long-term clinical benefit across subgroups in patients with KRAS p.G12C-mutated NSCLC, and additional biomarker data will be presented. Citation Format: Grace K. Dy, Ramaswamy Govindan, Vamsidhar Velcheti, Gerald S. Falchook, Antoine Italiano, Juergen Wolf, Adrian G. Sacher, Toshiaki Takahashi, Suresh S. Ramalingam, Christophe Dooms, Dong-Wan Kim, Alfredo Addeo, Jayesh Desai, Martin Schuler, Pascale Tomasini, Qui Tran, Simon Jones, Agnes Ang, Abraham Anderson, Antreas A. Hindoyan, David S. Hong, Bob T. Li. Long-term outcomes with sotorasib in pretreated KRASp.G12C-mutated NSCLC: 2-year analysis of CodeBreaK100 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT008.
- Published
- 2022
50. Five-year survival outcomes with nivolumab (NIVO) plus ipilimumab (IPI) versus chemotherapy (chemo) as first-line (1L) treatment for metastatic non–small cell lung cancer (NSCLC): Results from CheckMate 227
- Author
-
Julie R. Brahmer, Jong-Seok Lee, Tudor-Eliade Ciuleanu, Reyes Bernabe Caro, Makoto Nishio, Lazlo Urban, Clarisse Audigier-Valette, Lorena Lupinacci, Randeep S. Sangha, Luis G. Paz-Ares, Martin Reck, Hossein Borghaei, Kenneth John O'Byrne, Ravi Gupta, Judith Bushong, Li Li, Steven I. Blum, Laura Eccles, and Suresh S. Ramalingam
- Subjects
Cancer Research ,Oncology - Abstract
LBA9025 Background: In CheckMate 227 part 1 (NCT02477826), 1L NIVO + IPI demonstrated long-term, durable survival benefit vs platinum-doublet chemo in patients (pts) with metastatic NSCLC regardless of tumor programmed death ligand 1 (PD-L1) expression level. Here we present the longest reported follow-up (5 y) of a phase 3 trial of 1L combination immunotherapy in metastatic NSCLC. Methods: Adults with previously untreated stage IV or recurrent NSCLC, no known EGFR/ ALK alterations , and an ECOG performance status ≤ 1 were enrolled and stratified by histology. Pts with tumor PD-L1 ≥ 1% were randomized 1:1:1 to NIVO (3 mg/kg Q2W) + IPI (1 mg/kg Q6W), NIVO (240 mg Q2W), or chemo. Pts with tumor PD-L1 < 1% were randomized 1:1:1 to NIVO + IPI, NIVO (360 mg Q3W) + chemo, or chemo. Pts were treated until progression, toxicity, or ≤ 2 y for immunotherapy. Assessments included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), duration of response (DOR), and a novel efficacy endpoint, treatment-free interval. Treatment-free interval was measured in pts who discontinued study therapy (for any reason including treatment completion) and was defined as the time from last study dose to start of subsequent systemic therapy or death, whichever occurred first. Results: Minimum follow-up was 61.3 mo (database lock, Feb 15, 2022). In pts with tumor PD-L1 ≥ 1% (N = 1189), continued long-term OS benefit was seen with NIVO + IPI vs chemo (HR, 0.77 [95% CI, 0.66–0.91]); 5-y OS rates were 24% (NIVO + IPI), 17% (NIVO), and 14% (chemo). OS benefit also continued in pts with tumor PD-L1 < 1% (N = 550) for NIVO + IPI vs chemo (HR, 0.65 [95% CI, 0.52–0.81]); 5-y OS rates were 19% (NIVO + IPI), 10% (NIVO + chemo), and 7% (chemo). Clinical benefit with NIVO + IPI vs chemo was observed across additional efficacy endpoints in the overall population and in pts alive at 5 y (table). PFS, ORR, and DOR with NIVO and NIVO + chemo will be presented. Among pts alive at 5 y in the NIVO + IPI group, 66% (PD-L1 ≥ 1%) and 64% (PD-L1 < 1%) remained treatment-free ≥ 3 y after discontinuing study therapy; median (range) duration of NIVO ± IPI therapy was 17.7 (0-25.5) mo (PD-L1 ≥ 1%) and 9.5 (0-25.1) mo (PD-L1 < 1%). No new safety signals were observed. Conclusions: With a 5-y minimum follow-up, NIVO + IPI continues to provide long-term, durable clinical benefit vs chemo in previously untreated pts with metastatic NSCLC, regardless of PD-L1 expression. NIVO + IPI led to increased 5-y survivorship; the majority of these pts were treatment-free for ≥ 3 y post-treatment discontinuation. Clinical trial information: NCT02477826. [Table: see text]
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.