41 results on '"Konda B"'
Search Results
2. EP13.05-01 Early ctDNA Changes and Clinical Outcomes in Extensive Stage Small Cell Lung Cancer Patients on Nivolumab and Temozolomide
- Author
-
Gheeya, J.S., primary, Wei, L., additional, Bucheit, L., additional, Pilcher, C., additional, Sukrithan, V., additional, Konda, B., additional, Ferguson, S., additional, Jukich, M., additional, Savardekar, H., additional, Schwarz, E., additional, Norman, R., additional, Wesolowski, R., additional, Carson, W., additional, Kaufman, J., additional, Alahmadi, A., additional, Memmott, R., additional, Shields, P., additional, He, K., additional, Bertino, E., additional, Presley, C., additional, Carbone, D.P., additional, Verschraegen, C., additional, Otterson, G.A., additional, and Owen, D., additional
- Published
- 2023
- Full Text
- View/download PDF
3. Hypoxia and Cardiac Function in Patients With Prior Myocardial Infarction.
- Author
-
Hönemann, J.N., Gerlach, D., Hoffmann, F., Kramer, T., Weis, H., Hellweg, C.E., Konda, B., Zaha, V.G., Sadek, H.A., Herwaarden, A.E. van, Olthaar, A.J., Reuter, H., Baldus, S., Levine, B.D., Jordan, J., Tank, J., Limper, U., Hönemann, J.N., Gerlach, D., Hoffmann, F., Kramer, T., Weis, H., Hellweg, C.E., Konda, B., Zaha, V.G., Sadek, H.A., Herwaarden, A.E. van, Olthaar, A.J., Reuter, H., Baldus, S., Levine, B.D., Jordan, J., Tank, J., and Limper, U.
- Abstract
Item does not contain fulltext
- Published
- 2023
4. THE CELLULAR RESPONSE OF PRIMARY ASTROCYTES TO HEAVY ION EXPOSURE
- Author
-
Roggan, M.D., Kronenberg, J., Wollert, E., Hoffmann, S., Nisar, H., Konda, B., Diegeler, S., Liemersdorf, C., Hemmersbach, R., and Hellweg, C.E.
- Subjects
low- and high-LET radiation ,Primary murine astrocytes - Published
- 2023
5. EP14.05-004 Temozolomide and Atezolizumab as Second Line Treatment for Extensive Stage Small Cell Lung Cancer: A Randomized, Multi-Cohort Phase 2 Trial
- Author
-
Owen, D.H., primary, Durm, G.A., additional, Wei, L., additional, Pilcher, C., additional, Ferguson, S., additional, Benner, B., additional, Jukich, M., additional, Sukrithan, V., additional, Konda, B., additional, Savardekar, H., additional, Spakowicz, D., additional, Schwarz, E., additional, Norman, R.O., additional, Wesolowski, R., additional, Carson, W.E., additional, Kaufman, J., additional, Alahmadi, A., additional, Memmott, R., additional, Shields, P., additional, He, K., additional, Bertino, E.M., additional, Presley, C.J., additional, Carbone, D.P., additional, and Otterson, G.A., additional
- Published
- 2022
- Full Text
- View/download PDF
6. OA12.04 Efficacy of Nivolumab and Temozolomide in Extensive Stage Small Cell Lung Cancer after Chemo-Immunotherapy: A Phase 2 Trial.
- Author
-
Owen, D.H., primary, Wei, L., additional, Benner, B., additional, Pilcher, C., additional, Christenson, G., additional, Ferguson, S., additional, Jukich, M., additional, Sukrithan, V., additional, Konda, B., additional, Shah, M., additional, Savardekar, H., additional, Schwarz, E., additional, Norman, R.O., additional, Wesolowski, R., additional, Carson, W.E., additional, Kaufman, J., additional, Alahmadi, A., additional, Memmott, R., additional, Shields, P., additional, He, K., additional, Bertino, E.M., additional, Presley, C.J., additional, Carbone, D.P., additional, Verschraegen, C., additional, and Otterson, G.A., additional
- Published
- 2022
- Full Text
- View/download PDF
7. P3.13C.04 Assessing ctDNA using a Methylation-informed Molecular Response Algorithm in Extensive-Stage Small Cell Lung Cancer Patients
- Author
-
Gheeya, J.S., Bucheit, L., Solomon, S.R., Yablonovitch, A., Pilcher, C., Presley, C.J., Bertino, E., Shields, P.G., He, K., Sukrithan, V., Konda, B., Verschraegen, C.F., Carbone, D., Otterson, G.A., and Owen, D.H.
- Published
- 2024
- Full Text
- View/download PDF
8. 1141O Cabozantinib versus placebo for advanced neuroendocrine tumors (NET) after progression on prior therapy (CABINET Trial/Alliance A021602): Updated results including progression free-survival (PFS) by blinded independent central review (BICR) and subgroup analyses
- Author
-
Chan, J., Geyer, S., Zemla, T., Knopp, M.V., Behr, S.C., Pulsipher, S., Acoba, J., Shergill, A., Wolin, E.M., Halfdanarson, T.R., Konda, B., Trikalinos, N., Shaheen, S., Vijayvergia, N., Dasari, N.A., Strosberg, J., Kohn, E.C., Kulke, M., O'Reilly, E.M., and Meyerhardt, J.
- Published
- 2024
- Full Text
- View/download PDF
9. iPSC-Derived Alveolar Epithelial Type 2 Cells Resemble Their Primary Counterparts and Can Serve as Preclinical Disease Models of Pulmonary Diseases
- Author
-
Alysandratos, K.-D., primary, Garcia de Alba Rivas, C., additional, Yao, C., additional, Pessina, P., additional, Villacorta-Martin, C., additional, Huang, J., additional, Hix, O., additional, Konda, B., additional, Stripp, B.R., additional, Kim, C.F., additional, and Kotton, D.N., additional
- Published
- 2022
- Full Text
- View/download PDF
10. A randomized study to evaluate the safety and efficacy of two dosages of lenvatinib – 18 and 24 mg – in patients with radioiodrefract differentiated thyroid cancer
- Author
-
Brose, M. S., primary, Panaseykin, Yu., additional, Konda, B., additional, De la Fouchardiere, C., additional, Hughes, B. G.M., additional, Gianoukakis, A. G., additional, Park, Y. J., additional, Romanov, I., additional, Krzyzanowska, M. K., additional, Leboulleux, S., additional, Binder, T. A., additional, Dutcus, C., additional, Xie, R., additional, and Taylor, M. H., additional
- Published
- 2022
- Full Text
- View/download PDF
11. LBA53 Alliance A021602: Phase III, double-blinded study of cabozantinib versus placebo for advanced neuroendocrine tumors (NET) after progression on prior therapy (CABINET)
- Author
-
Chan, J., Geyer, S., Ou, F-S., Knopp, M., Behr, S., Zemla, T., Acoba, J., Shergill, A., Wolin, E.M., Halfdanarson, T.R., Trikalinos, N., Konda, B., Vijayvergia, N., Dasari, N.A., Strosberg, J., Kohn, E.C., Kulke, M.H., O'Reilly, E.M., and Meyerhardt, J.A.
- Published
- 2023
- Full Text
- View/download PDF
12. DNA Damage and Inflammatory Response of p53 Null H358 Non-Small Cell Lung Cancer Cells to X-Ray Exposure Under Chronic Hypoxia.
- Author
-
Nisar H, Brauny M, Labonté FM, Schmitz C, Konda B, and Hellweg CE
- Subjects
- Humans, X-Rays, Cell Line, Tumor, DNA Breaks, Double-Stranded radiation effects, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Cell Hypoxia, Gene Expression Regulation, Neoplastic radiation effects, Cell Survival radiation effects, Cell Survival genetics, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung radiotherapy, Tumor Suppressor Protein p53 metabolism, Tumor Suppressor Protein p53 genetics, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, DNA Damage
- Abstract
Hypoxia-induced radioresistance limits therapeutic success in cancer. In addition, p53 mutations are widespread in tumors including non-small cell lung carcinomas (NSCLCs), and they might modify the radiation response of hypoxic tumor cells. We therefore analyzed the DNA damage and inflammatory response in chronically hypoxic (1% O
2 , 48 h) p53 null H358 NSCLC cells after X-ray exposure. We used the colony-forming ability assay to determine cell survival, γH2AX immunofluorescence microscopy to quantify DNA double-strand breaks (DSBs), flow cytometry of DAPI-stained cells to measure cell cycle distribution, ELISAs to quantify IL-6 and IL-8 secretion in cell culture supernatants, and RNA sequencing to determine gene expression. Chronic hypoxia increased the colony-forming ability and radioresistance of H358 cells. It did not affect the formation or resolution of X-ray-induced DSBs. It reduced the fraction of cells undergoing G2 arrest after X-ray exposure and delayed the onset of G2 arrest. Hypoxia led to an earlier enhancement in cytokines secretion rate after X-irradiation compared to normoxic controls. Gene expression changes were most pronounced after the combined exposure to hypoxia and X-rays and pertained to senescence and different cell death pathways. In conclusion, hypoxia-induced radioresistance is present despite the absence of functional p53. This resistance is related to differences in clonogenicity, cell cycle regulation, cytokine secretion, and gene expression under chronic hypoxia, but not to differences in DNA DSB repair kinetics.- Published
- 2024
- Full Text
- View/download PDF
13. Inhibition of Bruton's tyrosine kinase with PD-1 blockade modulates T cell activation in solid tumors.
- Author
-
Schwarz E, Benner B, Wesolowski R, Quiroga D, Good L, Sun SH, Savardekar H, Li J, Jung KJ, Duggan MC, Lapurga G, Shaffer J, Scarberry L, Konda B, Verschraegen C, Kendra K, Shah M, Rupert R, Monk P, Shah HA, Noonan AM, Bixel K, Hays J, Wei L, Pan X, Behbehani G, Hu Y, Elemento O, Chung D, Xin G, Blaser BW, and Carson WE 3rd
- Subjects
- Humans, Male, Female, Middle Aged, Aged, T-Lymphocytes immunology, T-Lymphocytes drug effects, T-Lymphocytes metabolism, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Adult, Myeloid-Derived Suppressor Cells drug effects, Myeloid-Derived Suppressor Cells immunology, Myeloid-Derived Suppressor Cells metabolism, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors pharmacology, Pyrimidines therapeutic use, Pyrimidines pharmacology, Piperidines therapeutic use, Piperidines pharmacology, Agammaglobulinaemia Tyrosine Kinase antagonists & inhibitors, Agammaglobulinaemia Tyrosine Kinase metabolism, Adenine analogs & derivatives, Adenine therapeutic use, Adenine pharmacology, Neoplasms drug therapy, Neoplasms immunology, Lymphocyte Activation drug effects, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor metabolism, Nivolumab therapeutic use, Nivolumab pharmacology
- Abstract
BACKGROUNDInhibition of Bruton's tyrosine kinase with ibrutinib blocks the function of myeloid-derived suppressor cells (MDSC). The combination of ibrutinib and nivolumab was tested in patients with metastatic solid tumors.METHODSSixteen patients received ibrutinib 420 mg p.o. daily with nivolumab 240 mg i.v. on days 1 and 15 of a 28-day cycle. The effect of ibrutinib and nivolumab on MDSC, the immune profile, and cytokine levels were measured. Single-cell RNA-Seq and T cell receptor sequencing of immune cells was performed.RESULTSCommon adverse events were fatigue and anorexia. Four patients had partial responses and 4 had stable disease at 3 months (average 6.5 months, range 3.5-14.6). Median overall survival (OS) was 10.8 months. Seven days of Bruton's tyrosine kinase (BTK) inhibition significantly increased the proportion of monocytic-MDSC (M-MDSC) and significantly decreased chemokines associated with MDSC recruitment and accumulation (CCL2, CCL3, CCL4, CCL13). Single-cell RNA-Seq revealed ibrutinib-induced downregulation of genes associated with MDSC-suppressive function (TIMP1, CXCL8, VEGFA, HIF1A), reduced MDSC interactions with exhausted CD8+ T cells, and decreased TCR repertoire diversity. The addition of nivolumab significantly increased circulating NK and CD8+ T cells and increased CD8+ T cell proliferation. Exploratory analyses suggest that MDSC and T cell gene expression and TCR repertoire diversity were differentially affected by BTK inhibition according to patient response.CONCLUSIONIbrutinib and nivolumab were well tolerated and affected MDSC and T cell function in patients with solid metastatic tumors.TRIAL REGISTRATIONClinicalTrials.gov NCT03525925.FUNDINGNIH; National Cancer Institute Cancer; National Center for Advancing Translational Sciences; Pelotonia.
- Published
- 2024
- Full Text
- View/download PDF
14. Trends in the Use of Observation for Small Nonfunctional Pancreatic Neuroendocrine Tumors.
- Author
-
Ruff SM, Dillhoff M, Tsai S, Pawlik TM, Sukrithan V, Konda B, and Cloyd JM
- Published
- 2024
- Full Text
- View/download PDF
15. Phase 3 Trial of Cabozantinib to Treat Advanced Neuroendocrine Tumors.
- Author
-
Chan JA, Geyer S, Zemla T, Knopp MV, Behr S, Pulsipher S, Ou FS, Dueck AC, Acoba J, Shergill A, Wolin EM, Halfdanarson TR, Konda B, Trikalinos NA, Tawfik B, Raj N, Shaheen S, Vijayvergia N, Dasari A, Strosberg JR, Kohn EC, Kulke MH, O'Reilly EM, and Meyerhardt JA
- Abstract
Background: Treatment options for patients with advanced neuroendocrine tumors are limited. The efficacy of cabozantinib in the treatment of previously treated, progressive extrapancreatic or pancreatic neuroendocrine tumors is unclear., Methods: We enrolled two independent cohorts of patients - those with extrapancreatic neuroendocrine tumors and those with pancreatic neuroendocrine tumors - who had received peptide receptor radionuclide therapy or targeted therapy or both. Patients were randomly assigned in a 2:1 ratio to receive cabozantinib at a dose of 60 mg daily or placebo. The primary end point was progression-free survival as assessed by blinded independent central review. Key secondary end points included objective response, overall survival, and safety., Results: In the cohort of 203 patients with extrapancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 8.4 months, as compared with 3.9 months with placebo (stratified hazard ratio for progression or death, 0.38; 95% confidence interval [CI], 0.25 to 0.59; P<0.001). In the cohort of 95 patients with pancreatic neuroendocrine tumors, the median progression-free survival with cabozantinib was 13.8 months, as compared with 4.4 months with placebo (stratified hazard ratio, 0.23; 95% CI, 0.12 to 0.42; P<0.001). The incidence of confirmed objective response with cabozantinib was 5% and 19% among patients with extrapancreatic and pancreatic neuroendocrine tumors, respectively, as compared with 0% with placebo. Grade 3 or higher adverse events were noted in 62 to 65% of the patients treated with cabozantinib, as compared with 23 to 27% of the patients who received placebo. Common treatment-related adverse events of grade 3 or higher included hypertension, fatigue, diarrhea, and thromboembolic events., Conclusions: Cabozantinib, as compared with placebo, significantly improved progression-free survival in patients with previously treated, progressive advanced extrapancreatic or pancreatic neuroendocrine tumors. Adverse events were consistent with the known safety profile of cabozantinib. (Funded by the National Cancer Institute and others; CABINET ClinicalTrials.gov number, NCT03375320.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
16. Combination Targeted Therapy with Pembrolizumab and Lenvatinib in Progressive, Radioiodine-Refractory Differentiated Thyroid Cancers.
- Author
-
French JD, Haugen BR, Worden FP, Bowles DW, Gianoukakis AG, Konda B, Dadu R, Sherman EJ, McCue S, Foster NR, Nikiforov YE, Farias TDJ, Norman PJ, and Wirth LJ
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Protein Kinase Inhibitors therapeutic use, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Aged, 80 and over, Quinolines administration & dosage, Quinolines therapeutic use, Quinolines adverse effects, Phenylurea Compounds administration & dosage, Phenylurea Compounds therapeutic use, Phenylurea Compounds adverse effects, Thyroid Neoplasms drug therapy, Thyroid Neoplasms pathology, Thyroid Neoplasms mortality, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized adverse effects, Iodine Radioisotopes therapeutic use, Iodine Radioisotopes administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects
- Abstract
Purpose: Lenvatinib, a potent multikinase inhibitor, improves progression-free survival (PFS) in patients with radioiodine (RAI)-refractory differentiated thyroid cancer; however, most patients experience disease progression, warranting further therapy. We evaluated the efficacy and safety of lenvatinib plus pembrolizumab in these patients., Patients and Methods: We enrolled patients with progressive, RAI-refractory differentiated thyroid cancer who were either naïve to multikinase inhibitors (cohort 1) or who had progressed on lenvatinib (cohort 2). Patients received oral lenvatinib daily (cohort 1, 20 mg; cohort 2, dose at progression) and intravenous pembrolizumab (200 mg) every 21 days., Results: In cohorts 1 and 2, 30 and 27 patients were enrolled, respectively. Adverse events were consistent with those observed in other cancers. In cohort 1, the confirmed overall response rate was 65.5%. There were no complete responses (primary endpoint). The 12- and 18-month PFS were 72.0% and 58.0%, respectively, and the median PFS was 26.8 months. In cohort 2, the confirmed overall response rate was 16% (primary endpoint), and the median PFS was 10.0 months (95% confidence interval, 7.0-17.9 months). Tumor histology, driver mutations, and immune-related biomarkers, including PD-L1 expression, thyroid-specific antibody levels, and CD8+ T-cell tumor infiltrate, did not correlate with response to therapy. Increased baseline peripheral blood monocytes and neutrophil to lymphocyte ratio were associated with a worse PFS in cohort 1., Conclusions: Lenvatinib plus pembrolizumab may enhance the durability of lenvatinib monotherapy in lenvatinib-naïve patients. Furthermore, the addition of pembrolizumab may be a viable salvage therapy for patients who have progressed on lenvatinib., (©2024 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
17. Cabozantinib plus ipilimumab/nivolumab in patients with previously treated advanced differentiated thyroid cancer.
- Author
-
Konda B, Sherman EJ, Massarelli E, Nieva J, Muzaffar J, Morris Iii JC, Ryder M, Ho AL, Agulnik M, Wei L, Handley D, Moses C, Jacob R, Wright J, Streicher H, Carson W, and Shah MH
- Abstract
Background: This investigator-initiated phase II trial aimed to evaluate the efficacy of cabozantinib in combination with nivolumab and ipilimumab (CaboNivoIpi) in previously treated patients with radioactive iodine (RAI)-refractory differentiated thyroid cancer (DTC) (NCT03914300)., Methods: Eligible patients with RAI-refractory DTC who progressed on 1 prior line of VEGFR-targeted therapy received a 2-week run-in of cabozantinib monotherapy followed by CaboNivoIpi for 4 cycles (cycle length = 6 weeks), followed by cabozantinib plus nivolumab (cycle length = 4 weeks) until disease progression. The primary endpoint was objective response rate (ORR) within the first 6 months of treatment. A Simon optimal 2-stage design allowed for an interim analysis after accrual of 10 evaluable patients. At least 5 responses were needed to proceed to stage 2., Results: Among 11 patients enrolled, the median age was 69 years. Prior VEGFR-targeted therapies included lenvatinib, pazopanib, and sorafenib plus everolimus. Median follow-up was 7.9 months. Among 10 evaluable patients, ORR within the first 6 months of treatment was 10% (1 partial response). Median progression-free survival was 9 months [95% CI: 3.0, not reached] and median overall survival was 19.2 months [(95% CI: 4.6, not reached]. Grade 3/4 treatment-related adverse events (AEs) were noted in 55% (6/11) and grade 5 AEs in 18% (2/11) of patients. The most common treatment-related AE was hypertension. The study did not reach its prespecified efficacy threshold., Conclusion: CaboNivoIpi had low ORRs and a high rate of grade ≥3 treatment-related AEs., Clinical Trial Registration: NCT03914300., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com. See the journal About page for additional terms.)
- Published
- 2024
- Full Text
- View/download PDF
18. Safety and efficacy of peptide receptor radionuclide therapy in neuroendocrine tumors: A single center experience.
- Author
-
Sukrithan V, Armbruster H, Rogers S, Vogt SM, Grenade C, Verschraegen C, Zhou Y, Goyal A, Natwa M, Hussein A, Barr H, Konate D, Batdorf R, Brown A, Williams B, Zhao S, Wei L, Xu M, Shah MH, and Konda B
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Treatment Outcome, Organometallic Compounds therapeutic use, Organometallic Compounds adverse effects, Organometallic Compounds administration & dosage, Aged, 80 and over, Radiopharmaceuticals therapeutic use, Radiopharmaceuticals adverse effects, Radiopharmaceuticals administration & dosage, Pancreatic Neoplasms radiotherapy, Pancreatic Neoplasms pathology, Retrospective Studies, Neuroendocrine Tumors radiotherapy, Neuroendocrine Tumors pathology, Neuroendocrine Tumors metabolism, Octreotide analogs & derivatives, Octreotide therapeutic use, Octreotide adverse effects, Octreotide administration & dosage, Receptors, Peptide metabolism
- Abstract
Peptide receptor radionucleotide therapy (PRRT) with 177Lu-dotatate is widely used for the treatment of patients with neuroendocrine tumors (NETs). We analyzed data from 104 patients with NETs treated with 177Lu -dotatate at a US academic center between December 2017 and October 2020 to better understand patterns of long-term efficacy, safety, and toxicity in the real-world setting. 177Lu-dotatate (200 mCi) was administered every eight weeks for four doses. The most common sites of primary disease were small intestine NETs (n = 49, 47%), pancreatic NETs (n = 32, 31%), and lung NETs (n = 7, 7%). Twenty-seven percent had Ki-67 <3%, 49% had Ki-67 between 3-20%, and 13.5% had Ki-67 >20%. The cohort had been pretreated with a median of two prior lines of treatment. Forty percent had received prior liver-directed treatment. Seventy-four percent of patients completed all four doses of treatment. The objective response rate was 18%. The median time-to-treatment failure/death was significantly longer for small-bowel NETs when compared to pancreatic NETs (37.3 months vs. 13.2 months, p = 0.001). In a multivariate model, Ki-67, primary site, and liver tumor burden ≥50% were found to independently predict time-to-treatment failure/death. Around 40% of patients experienced adverse events of ≥grade 3 severity. Treatment-related adverse events leading to discontinuation of therapy happened in 10% of patients. Preexisting mesenteric/peritoneal disease was present in 33 patients; seven of these patients developed bowel-related toxicities including two grade 5 events. We also report two cases of delayed-onset minimal change nephrotic syndrome, which occurred 14 and 27 months after the last dose of PRRT. Lastly, we describe six patients who developed rapid tumor progression in the liver leading to terminal liver failure within 7.3 months from the start of PRRT, and identify potential risk factors associated with this occurrence, which will need further study., Competing Interests: The authors have read the journal’s policy and have the following competing interests: Vineeth Sukrithan has received clinical research, on behalf of Ohio State University, funding from Eli Lilly and Company and Crinetics Pharmaceuticals, as well as consulting fees from GE Oncology, Exelixis, and Lantheus. Bhavana Konda has received grant funding, on behalf of Ohio State University, from Xencor, Merck, and Eisai., (Copyright: © 2024 Sukrithan et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2024
- Full Text
- View/download PDF
19. Clinical Activity of Selpercatinib in RET-mutant Pheochromocytoma.
- Author
-
Deschler-Baier B, Konda B, Massarelli E, Hu MI, Wirth LJ, Xu X, Wright J, and Clifton-Bligh RJ
- Abstract
Introduction: Activating RET alterations have been reported in a variety of solid tumors, including pheochromocytoma where they occur both sporadically and as part of familial multiple endocrine neoplasia type 2 (MEN2) syndromes. Selpercatinib is a first-in-class, highly selective, and potent small molecule RET kinase inhibitor that has demonstrated marked and durable anti-tumor activity in diverse RET-activated solid tumors in the LIBRETTO-001 study (NCT03157128)., Methods: We describe the first six pheochromocytoma cases treated with selpercatinib in the LIBRETTO-001 study., Results: Of the six patients (one sporadic and five reported as part of MEN2 syndromes) in this case report, four had a partial response/complete response and two had stable disease per independent review committee. Treatment duration ranged from 9.2 months to more than 56.4 months. The safety profile of treatment was consistent with selpercatinib in other indications., Conclusion: These data support selpercatinib as an effective therapy against RET-mutant pheochromocytoma, adding to the diversity of RET-activated tumor types that may benefit from targeted RET inhibition., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2024
- Full Text
- View/download PDF
20. NF-κB in the Radiation Response of A549 Non-Small Cell Lung Cancer Cells to X-rays and Carbon Ions under Hypoxia.
- Author
-
Nisar H, Sanchidrián González PM, Labonté FM, Schmitz C, Roggan MD, Kronenberg J, Konda B, Chevalier F, and Hellweg CE
- Subjects
- Humans, A549 Cells, X-Rays, Gene Expression Regulation, Neoplastic radiation effects, Linear Energy Transfer, Cell Hypoxia radiation effects, Carbon, Cell Survival radiation effects, Radiation Tolerance, Interleukin-8 metabolism, Interleukin-8 genetics, NF-kappa B metabolism, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung radiotherapy, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms metabolism, Lung Neoplasms radiotherapy, Lung Neoplasms pathology, Lung Neoplasms genetics
- Abstract
Cellular hypoxia, detectable in up to 80% of non-small cell lung carcinoma (NSCLC) tumors, is a known cause of radioresistance. High linear energy transfer (LET) particle radiation might be effective in the treatment of hypoxic solid tumors, including NSCLC. Cellular hypoxia can activate nuclear factor κB (NF-κB), which can modulate radioresistance by influencing cancer cell survival. The effect of high-LET radiation on NF-κB activation in hypoxic NSCLC cells is unclear. Therefore, we compared the effect of low (X-rays)- and high (
12 C)-LET radiation on NF-κB responsive genes' upregulation, as well as its target cytokines' synthesis in normoxic and hypoxic A549 NSCLC cells. The cells were incubated under normoxia (20% O2 ) or hypoxia (1% O2 ) for 48 h, followed by irradiation with 8 Gy X-rays or12 C ions, maintaining the oxygen conditions until fixation or lysis. Regulation of NF-κB responsive genes was evaluated by mRNA sequencing. Secretion of NF-κB target cytokines, IL-6 and IL-8, was quantified by ELISA. A greater fold change increase in expression of NF-κB target genes in A549 cells following exposure to12 C ions compared to X-rays was observed, regardless of oxygenation status. These genes regulate cell migration, cell cycle, and cell survival. A greater number of NF-κB target genes was activated under hypoxia, regardless of irradiation status. These genes regulate cell migration, survival, proliferation, and inflammation. X-ray exposure under hypoxia additionally upregulated NF-κB target genes modulating immunosurveillance and epithelial-mesenchymal transition (EMT). Increased IL-6 and IL-8 secretion under hypoxia confirmed NF-κB-mediated expression of pro-inflammatory genes. Therefore, radiotherapy, particularly with X-rays, may increase tumor invasiveness in surviving hypoxic A549 cells., Competing Interests: The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.- Published
- 2024
- Full Text
- View/download PDF
21. Hypoxia Modulates Radiosensitivity and Response to Different Radiation Qualities in A549 Non-Small Cell Lung Cancer (NSCLC) Cells.
- Author
-
Nisar H, Labonté FM, Roggan MD, Schmitz C, Chevalier F, Konda B, Diegeler S, Baumstark-Khan C, and Hellweg CE
- Subjects
- Humans, A549 Cells, Hypoxia, Radiation Tolerance, Oxygen, Ions, Phosphatidylinositol 3-Kinases, Tumor Microenvironment, Carcinoma, Non-Small-Cell Lung radiotherapy, Lung Neoplasms radiotherapy
- Abstract
Hypoxia-induced radioresistance reduces the efficacy of radiotherapy for solid malignancies, including non-small cell lung cancer (NSCLC). Cellular hypoxia can confer radioresistance through cellular and tumor micro-environment adaptations. Until recently, studies evaluating radioresistance secondary to hypoxia were designed to maintain cellular hypoxia only before and during irradiation, while any handling of post-irradiated cells was carried out in standard oxic conditions due to the unavailability of hypoxia workstations. This limited the possibility of simulating in vivo or clinical conditions in vitro. The presence of molecular oxygen is more important for the radiotoxicity of low-linear energy transfer (LET) radiation (e.g., X-rays) than that of high-LET carbon (
12 C) ions. The mechanisms responsible for12 C ions' potential to overcome hypoxia-induced radioresistance are currently not fully understood. Therefore, the radioresistance of hypoxic A549 NSCLC cells following exposure to X-rays or12 C ions was investigated along with cell cycle progression and gene expression by maintaining hypoxia before, during and after irradiation. A549 cells were incubated under normoxia (20% O2 ) or hypoxia (1% O2 ) for 48 h and then irradiated with X-rays (200 kV) or12 C ions (35 MeV/n, LET ~75 keV/µm). Cell survival was evaluated using colony-forming ability (CFA) assays immediately or 24 h after irradiation (late plating). DNA double-strand breaks (DSBs) were analyzed using γH2AX immunofluorescence microscopy. Cell cycle progression was determined by flow cytometry of 4',6-diamidino-2-phenylindole-stained cells. The global transcription profile post-irradiation was evaluated by RNA sequencing. When hypoxia was maintained before, during and after irradiation, hypoxia-induced radioresistance was observed only in late plating CFA experiments. The killing efficiency of12 C ions was much higher than that of X-rays. Cell survival under hypoxia was affected more strongly by the timepoint of plating in the case of X-rays compared to12 C ions. Cell cycle arrest following irradiation under hypoxia was less pronounced but more prolonged. DSB induction and resolution following irradiation were not significantly different under normoxia and hypoxia. Gene expression response to irradiation primarily comprised cell cycle regulation for both radiation qualities and oxygen conditions. Several PI3K target genes involved in cell migration and cell motility were differentially upregulated in hypoxic cells. Hypoxia-induced radioresistance may be linked to altered cell cycle response to irradiation and PI3K-mediated changes in cell motility and migration in A549 cells rather than less DNA damage or faster repair.- Published
- 2024
- Full Text
- View/download PDF
22. Neoadjuvant dabrafenib and trametinib for functional organ preservation in recurrent BRAF V600E-mutated papillary thyroid cancer.
- Author
-
Farlow JL, McCrary HC, Sipos JA, Phay JE, Konda B, and Agrawal A
- Subjects
- Female, Humans, Thyroid Cancer, Papillary drug therapy, Neoadjuvant Therapy, Organ Preservation, Pyridones pharmacology, Pyridones therapeutic use, Mutation, Antineoplastic Combined Chemotherapy Protocols, Proto-Oncogene Proteins B-raf genetics, Thyroid Neoplasms drug therapy, Thyroid Neoplasms genetics
- Abstract
Objectives: To describe the first reported use of neoadjuvant dabrafenib and trametinib specifically to permit organ conservation surgery in locally advanced recurrent differentiated thyroid carcinoma., Patients and Methods: A patient presented with locally recurrent, radioiodide-resistant DTC with a BRAF V600E mutation invading the laryngotrachea. Definitive treatment would require a total laryngectomy. She was offered neoadjuvant dabrafenib and trametinib prior to surgery., Results: A significant radiographic response permitted partial laryngectomy, enabling preservation of voice, early resumption of oral feeding, and avoidance of permanent tracheostomy. At 9 months, she remained free of disease., Conclusion: Neoadjuvant tyrosine kinase inhibitor treatment prior to definitive surgery for locally-invasive recurrent DTC is a potential approach that may limit the degree of surgery and associated morbidity., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
23. Coronary Artery and Peripheral Vascular Disease in a Patient with Poorly Differentiated Thyroid Cancer Treated with the Tyrosine Kinase Inhibitor Lenvatinib.
- Author
-
Sukrithan V, Kim L, Sipos JA, Goyal A, Zhou Y, Addison D, Shah M, Konda B, and Vallakati A
- Abstract
A subset of patients with differentiated thyroid carcinoma develop radioiodine refractory (RAIR) incurable disease, which typically has a poor prognosis. The multitargeted tyrosine kinase inhibitor lenvatinib has demonstrated significant improvements in progression-free survival in RAIR thyroid cancers compared to placebos. However, in the phase III SELECT trial of the drug in thyroid cancer, 5.4% of patients on lenvatinib experienced arterial thromboembolic events, with 2.7% experiencing severe grade ≥3 toxicities associated with arterial vascular events. This case study reports a patient with metastatic poorly differentiated follicular thyroid cancer who developed significant obstructive coronary artery disease following initiation of lenvatinib treatment, despite no predisposing cardiovascular risk factors apart from a remote smoking history. The possibility of developing coronary or peripheral artery disease should be considered in patients who are on targeted therapies, such as lenvatinib, even in the absence of traditional cardiovascular risk factors. In addition, baseline cardiac risk assessment and early treatment should be pursued to minimize interruptions to potentially lifesaving cancer therapy., Competing Interests: The authors declare that they have no conflicts of interest., (Copyright © 2023 Vineeth Sukrithan et al.)
- Published
- 2023
- Full Text
- View/download PDF
24. Secondary prevention of cardiovascular disease: unrecognized opportunity to improve survival in cancer patients.
- Author
-
Vallakati A and Konda B
- Subjects
- Humans, Secondary Prevention, Behavioral Risk Factor Surveillance System, Cardiovascular Diseases diagnosis, Cardiovascular Diseases epidemiology, Cardiovascular Diseases prevention & control, Neoplasms diagnosis, Neoplasms epidemiology, Neoplasms prevention & control, Cardiovascular System
- Abstract
Competing Interests: Conflict of interest: None declared.
- Published
- 2023
- Full Text
- View/download PDF
25. Clinical Outcomes of DEB-TACE in Hepatic Metastatic Neuroendocrine Tumors: A 5-Year Single-Institutional Experience.
- Author
-
Makary MS, Regalado LE, Alexander J, Sukrithan V, Konda B, and Cloyd JM
- Subjects
- Humans, Male, Female, Middle Aged, Retrospective Studies, Treatment Outcome, Doxorubicin, Liver Neoplasms diagnostic imaging, Liver Neoplasms therapy, Liver Neoplasms pathology, Carcinoma, Hepatocellular therapy, Neuroendocrine Tumors diagnostic imaging, Neuroendocrine Tumors therapy, Chemoembolization, Therapeutic
- Abstract
Rationale and Objectives: To evaluate single-institution outcomes of drug-eluting bead transarterial chemoembolization (DEB-TACE) in the treatment of locally advanced neuroendocrine tumor (NET) hepatic metastases with a focus on safety and efficacy of treatment., Materials and Methods: A single-center retrospective cohort study of the outcomes of consecutive patients with NELM who underwent DEB-TACE between 2014 and 2019 was performed. Clinicopathologic characteristics, radiologic response (modified Response Evaluation Criteria in Solid Tumors) at 1-month follow-up, adverse events, progression-free survival (PFS), and overall survival were calculated., Results: Among 287 patients (mean age of 62 years; 39% male: 61% female), disease burden was bilobar (90.2%) with mean largest tumor diameter measuring 4.9 ± 2.8 cm. Of these patients, 14.6% had no evidence of tumor in other organs or lymph nodes. Complete response occurred in 60 (20.9%) patients while 133 (46.3%) had partial responses. Major complication occurred in 2.4%. Liver function tests including total bilirubin and AST were overall stable at the 1-month follow-up, with only a small increase in the ALT at +8.9% (p < 0.01). Overall survival was 80.1% at 1 year, 49.1% at 3 years, and 12.3% at 5 years with a mean PFS of 14.4 ± 12.5 months., Conclusion: Based on this institutional experience, DEB-TACE is an acceptable locoregional therapy choice for hepatic metastases of NET due to its tolerable safety profile and relative efficacy., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Association of University Radiologists. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
26. Comparative Analysis of Molecular Pathogenic Mechanisms and Antiviral Development Targeting Old and New World Hantaviruses.
- Author
-
Jeyachandran AV, Irudayam JI, Dubey S, Chakravarty N, Konda B, Shah A, Su B, Wang C, Cui Q, Williams KJ, Srikanth S, Shi Y, Deb A, Damoiseaux R, Stripp BR, Ramaiah A, and Arumugaswami V
- Abstract
Background: Hantaviruses - dichotomized into New World (i.e. Andes virus, ANDV; Sin Nombre virus, SNV) and Old-World viruses (i.e. Hantaan virus, HTNV) - are zoonotic viruses transmitted from rodents to humans. Currently, no FDA-approved vaccines against hantaviruses exist. Given the recent breakthrough to human-human transmission by the ANDV, an essential step is to establish an effective pandemic preparedness infrastructure to rapidly identify cell tropism, infective potential, and effective therapeutic agents through systematic investigation., Methods: We established human cell model systems in lung (airway and distal lung epithelial cells), heart (pluripotent stem cell-derived (PSC-) cardiomyocytes), and brain (PSC-astrocytes) cell types and subsequently evaluated ANDV, HTNV and SNV tropisms. Transcriptomic, lipidomic and bioinformatic data analyses were performed to identify the molecular pathogenic mechanisms of viruses in different cell types. This cell-based infection system was utilized to establish a drug testing platform and pharmacogenomic comparisons., Results: ANDV showed broad tropism for all cell types assessed. HTNV replication was predominantly observed in heart and brain cells. ANDV efficiently replicated in human and mouse 3D distal lung organoids. Transcriptomic analysis showed that ANDV infection resulted in pronounced inflammatory response and downregulation of cholesterol biosynthesis pathway in lung cells. Lipidomic profiling revealed that ANDV-infected cells showed reduced level of cholesterol esters and triglycerides. Further analysis of pathway-based molecular signatures showed that, compared to SNV and HTNV, ANDV infection caused drastic lung cell injury responses. A selective drug screening identified STING agonists, nucleoside analogues and plant-derived compounds that inhibited ANDV viral infection and rescued cellular metabolism. In line with experimental results, transcriptome data shows that the least number of total and unique differentially expressed genes were identified in urolithin B- and favipiravir-treated cells, confirming the higher efficiency of these two drugs in inhibiting ANDV, resulting in host cell ability to balance gene expression to establish proper cell functioning., Conclusions: Overall, our study describes advanced human PSC-derived model systems and systems-level transcriptomics and lipidomic data to better understand Old and New World hantaviral tropism, as well as drug candidates that can be further assessed for potential rapid deployment in the event of a pandemic., Competing Interests: Competing interests. The authors declare no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
27. Maladaptive TGF-β Signals to the Alveolar Epithelium Drive Fibrosis after COVID-19 Infection.
- Author
-
Yao C, Parimon T, Espindola MS, Hohmann MS, Konda B, Hogaboam CM, Stripp BR, and Chen P
- Subjects
- Humans, Transforming Growth Factor beta, Fibrosis, Epithelium, Transforming Growth Factor beta1, COVID-19, Pulmonary Fibrosis etiology
- Published
- 2023
- Full Text
- View/download PDF
28. Selpercatinib for adult patients with locally advanced or metastatic RET-altered solid tumors.
- Author
-
Le D and Konda B
- Subjects
- Adult, Proto-Oncogene Proteins c-ret genetics, Protein Kinase Inhibitors therapeutic use, Humans, Carcinoma, Neuroendocrine, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Thyroid Neoplasms drug therapy, Thyroid Neoplasms genetics, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics
- Abstract
Introduction: The rearranged during transfection (RET) mutation is an oncogene driver for the development of cancer. Selpercatinib is a highly selective RET inhibitor that has demonstrated anti-tumor activity in RET-mutated cancers. Selpercatinib is approved for use in RET fusion-positive non-small-cell lung cancer (NSCLC), RET-mutated medullary thyroid cancer, RET fusion-positive thyroid cancer, and RET fusion-positive solid tumors., Areas Covered: This review summarizes the pharmacology, efficacy, safety, and resistance mechanism of selpercatinib., Expert Opinion: Selpercatinib has demonstrated durable responses with a favorable safety profile making it an excellent treatment option for RET-mutated cancers. Clinical trials are currently underway to determine the optimal sequencing of selpercatinib in RET fusion-positive lung and RET-mutated medullary thyroid cancer in the first-line setting compared to the current standard of care. Selpercatinib has shown promising anti-tumor activity in various RET-altered solid tumors opening a new treatment option for these patients.
- Published
- 2023
- Full Text
- View/download PDF
29. Hypoxia and Cardiac Function in Patients With Prior Myocardial Infarction.
- Author
-
Hönemann JN, Gerlach D, Hoffmann F, Kramer T, Weis H, Hellweg CE, Konda B, Zaha VG, Sadek HA, van Herwarden AE, Olthaar AJ, Reuter H, Baldus S, Levine BD, Jordan J, Tank J, and Limper U
- Subjects
- Humans, Hypoxia, Altitude, Myocardial Infarction
- Published
- 2023
- Full Text
- View/download PDF
30. Hypoxia Changes Energy Metabolism and Growth Rate in Non-Small Cell Lung Cancer Cells.
- Author
-
Nisar H, Sanchidrián González PM, Brauny M, Labonté FM, Schmitz C, Roggan MD, Konda B, and Hellweg CE
- Abstract
Hypoxia occurs in 80% of non-small cell lung carcinoma (NSCLC) cases, leading to treatment resistance. Hypoxia's effects on NSCLC energetics are not well-characterized. We evaluated changes in glucose uptake and lactate production in two NSCLC cell lines under hypoxia in conjunction with growth rate and cell cycle phase distribution. The cell lines A549 (p53 wt) and H358 (p53 null) were incubated under hypoxia (0.1% and 1% O
2 ) or normoxia (20% O2 ). Glucose and lactate concentrations in supernatants were measured using luminescence assays. Growth kinetics were followed over seven days. Cell nuclei were stained with DAPI and nuclear DNA content was determined by flow cytometry to determine cell cycle phase. Gene expression under hypoxia was determined by RNA sequencing. Glucose uptake and lactate production under hypoxia were greater than under normoxia. They were also significantly greater in A549 compared to H358 cells. Faster energy metabolism in A549 cells was associated with a higher growth rate in comparison to H358 cells under both normoxia and hypoxia. In both cell lines, hypoxia significantly slowed down the growth rate compared to proliferation under normoxic conditions. Hypoxia led to redistribution of cells in the different cycle phases: cells in G1 increased and the G2 population decreased. Glucose uptake and lactate production increase under hypoxia in NSCLC cells indicated greater shunting of glucose into glycolysis rather than into oxidative phosphorylation compared to normoxia, making adenosine triphosphate (ATP) production less efficient. This may explain the redistribution of hypoxic cells in the G1 cell cycle phase and the time increase for cell doubling. Energy metabolism changes were more prominent in faster-growing A549 cells compared to slower-growing H358 cells, indicating possible roles for the p53 status and inherent growth rate of different cancer cells. In both cell lines, genes associated with cell motility, locomotion and migration were upregulated under chronic hypoxia, indicating a strong stimulus to escape hypoxic conditions.- Published
- 2023
- Full Text
- View/download PDF
31. Unraveling astrocyte behavior in the space brain: Radiation response of primary astrocytes.
- Author
-
Roggan MD, Kronenberg J, Wollert E, Hoffmann S, Nisar H, Konda B, Diegeler S, Liemersdorf C, and Hellweg CE
- Subjects
- Animals, Mice, Interleukin-6, Ions, Brain, RNA, Messenger, DNA, Astrocytes metabolism, NF-kappa B
- Abstract
Introduction: Exposure to space conditions during crewed long-term exploration missions can cause several health risks for astronauts. Space radiation, isolation and microgravity are major limiting factors. The role of astrocytes in cognitive disturbances by space radiation is unknown. Astrocytes' response toward low linear energy transfer (LET) X-rays and high-LET carbon (
12 C) and iron (56 Fe) ions was compared to reveal possible effects of space-relevant high-LET radiation. Since astronauts are exposed to ionizing radiation and microgravity during space missions, the effect of simulated microgravity on DNA damage induction and repair was investigated., Methods: Primary murine cortical astrocytes were irradiated with different doses of X-rays,12 C and56 Fe ions at the heavy ion accelerator GSI. DNA damage and repair (γH2AX, 53BP1), cell proliferation (Ki-67), astrocytes' reactivity (GFAP) and NF-κB pathway activation (p65) were analyzed by immunofluorescence microscopy. Cell cycle progression was investigated by flow cytometry of DNA content. Gene expression changes after exposure to X- rays were investigated by mRNA-sequencing. RT-qPCR for several genes of interest was performed with RNA from X-rays- and heavy-ion-irradiated astrocytes: Cdkn1a, Cdkn2a, Gfap, Tnf , Il1 β, Il6 , and Tgf β 1 . Levels of the pro inflammatory cytokine IL-6 were determined using ELISA. DNA damage response was investigated after exposure to X-rays followed by incubation on a 2D clinostat to simulate the conditions of microgravity., Results: Astrocytes showed distinct responses toward the three different radiation qualities. Induction of radiation-induced DNA double strand breaks (DSBs) and the respective repair was dose-, LET- and time-dependent. Simulated microgravity had no significant influence on DNA DSB repair. Proliferation and cell cycle progression was not affected by radiation qualities examined in this study. Astrocytes expressed IL-6 and GFAP with constitutive NF-κB activity independent of radiation exposure. mRNA sequencing of X-irradiated astrocytes revealed downregulation of 66 genes involved in DNA damage response and repair, mitosis, proliferation and cell cycle regulation., Discussion: In conclusion, primary murine astrocytes are DNA repair proficient irrespective of radiation quality. Only minor gene expression changes were observed after X-ray exposure and reactivity was not induced. Co-culture of astrocytes with microglial cells, brain organoids or organotypic brain slice culture experiments might reveal whether astrocytes show a more pronounced radiation response in more complex network architectures in the presence of other neuronal cell types., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Roggan, Kronenberg, Wollert, Hoffmann, Nisar, Konda, Diegeler, Liemersdorf and Hellweg.)- Published
- 2023
- Full Text
- View/download PDF
32. A Phase II Clinical Trial of Nivolumab and Temozolomide for Neuroendocrine Neoplasms.
- Author
-
Owen DH, Benner B, Wei L, Sukrithan V, Goyal A, Zhou Y, Pilcher C, Suffren SA, Christenson G, Curtis N, Jukich M, Schwarz E, Savardekar H, Norman R, Ferguson S, Kleiber B, Wesolowski R, Carson WE, Otterson GA, Verschraegen CF, Shah MH, and Konda B
- Subjects
- Humans, Nivolumab therapeutic use, Temozolomide therapeutic use, Progression-Free Survival, Lung Neoplasms drug therapy, Neuroendocrine Tumors drug therapy
- Abstract
Purpose: Treatment options are limited in patients with metastatic neuroendocrine neoplasms (NEN). We present the results for a phase II trial of combination nivolumab and temozolomide in patients with advanced NEN along with results of immune changes in peripheral blood., Patients and Methods: NCT03728361 is a nonrandomized, phase II study of nivolumab and temozolomide in patients with NEN. The primary endpoint was response rate using RECIST 1.1. Secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety. Immune profiling was performed by mass cytometry to evaluate the effect on peripheral blood immune cell subsets., Results: Among all 28 patients with NEN, the confirmed response rate was 9/28 [32.1%, 95% confidence interval (CI): 15.9-52.4]. Of 11 patients with lung NEN, the response rate was 64% (n = 7); there was a significant difference in responses by primary tumor location (lung vs. others, P = 0.020). The median PFS was 8.8 months (95% CI: 3.9-11.1 months), and median OS was 32.3 months (95% CI: 20.7-not reached months). Exploratory blood immune cell profiling revealed an increase in circulating CD8+ T cells (27.9% ± 13.4% vs. 31.7% ± 14.6%, P = 0.03) and a decrease in CD4+ T cells (59.6% ± 13.1% vs. 56.5% ± 13.0%, P = 0.001) after 2 weeks of treatment. LAG-3-expressing total T cells were lower in patients experiencing a partial response (0.18% ± 0.24% vs. 0.83% ± 0.55%, P = 0.028). Myeloid-derived suppressor cell levels increased during the study and did not correlate with response., Conclusions: Combination nivolumab and temozolomide demonstrated promising activity in NEN. See related commentary by Velez and Garon, p. 691., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
33. Health-related quality-of-life analyses from a multicenter, randomized, double-blind phase 2 study of patients with differentiated thyroid cancer treated with lenvatinib 18 or 24 mg/day.
- Author
-
Taylor MH, Leboulleux S, Panaseykin Y, Konda B, de La Fouchardiere C, Hughes BGM, Gianoukakis AG, Park YJ, Romanov I, Krzyzanowska MK, Garbinsky D, Sherif B, Pan JJ, Binder TA, Sauter N, Xie R, and Brose MS
- Subjects
- Humans, Iodine Radioisotopes therapeutic use, Double-Blind Method, Quality of Life, Thyroid Neoplasms drug therapy, Adenocarcinoma drug therapy
- Abstract
Background: In the phase 2 double-blind Study 211, a starting dose of lenvatinib 18 mg/day was compared with the approved starting dose of 24 mg/day in patients with radioiodine-refractory differentiated thyroid cancer (RR-DTC). Predefined criteria for noninferiority for efficacy in the 18 mg arm were not met; safety was similar in both arms. Impact of lenvatinib treatment on health-related quality-of-life (HRQoL) was a secondary endpoint of Study 211., Methods: Patients with RR-DTC were randomly assigned to a blinded starting dose of lenvatinib 18 mg/day or 24 mg/day. HRQoL was assessed at baseline, every 8 weeks until Week 24, then every 16 weeks, and at the off-treatment visit, using the EQ-5D-3L and FACT-G instruments. Completion and compliance rates, mean change from baseline, and times to first and definitive deterioration were evaluated., Results: Baseline EQ-5D and FACT-G scores, and overall changes from baseline, were comparable between patients in the lenvatinib 18 mg/day (n = 77) and 24 mg/day arms (n = 75). For the 18 mg versus 24 mg arms, least squares mean differences were -0.42 (95% CI -4.88, 4.03) for EQ-5D-VAS and 0.47 (95% CI -3.45, 4.39) for FACT-G total. Time to first deterioration did not significantly favor either arm; EQ-5D-VAS HR [18 mg/24 mg] 0.93 (95% CI 0.61-1.40), EQ-5D-HUI HR [18 mg/24 mg] 0.68 (95% CI 0.44-1.05), FACT-G total HR [18 mg/24 mg] 0.73 (95% CI 0.48-1.12). Time to definitive deterioration did not significantly favor either arm, though EQ-5D-VAS showed a trend in favor of the 24 mg arm (HR [18 mg/24 mg] 1.72; 95% CI 0.99-3.01); EQ-5D-HUI HR [18 mg/24 mg] was 0.96 (95% CI 0.57-1.63), FACT-G total HR [18 mg/24 mg] was 0.72 (95% CI 0.43-1.21)., Conclusions: In Study 211, HRQoL for patients in the lenvatinib 18 mg/day arm was not statistically different from that of patients in the 24 mg/day arm. These data further support the use of the approved lenvatinib starting dose of 24 mg/day in patients with RR-DTC., Gov Number: NCT02702388., (© 2022 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
34. Kinase inhibitors in thyroid cancers.
- Author
-
Sukrithan V, Jain P, Shah MH, and Konda B
- Abstract
Objective: The treatment landscape for thyroid cancers has changed rapidly with the availability of kinase inhibitors against VEGFR, BRAF, MEK, NTRK, and RET. We provide an up-to-date review of the role of kinase inhibitors in thyroid cancer and discuss upcoming trials., Design & Methods: A comprehensive review of the available literature describing kinase inhibitors in thyroid cancer was performed., Results and Conclusions: Kinase inhibitors have become the standard of care for patients with metastatic radioactive iodine-refractory thyroid cancer. Short-term treatment can re-sensitize differentiated thyroid cancer to radioactive iodine, thereby potentially improving outcomes and sparing toxicities associated with the long-term use of kinase inhibitors. The approval of cabozantinib as salvage therapy for progressive radioactive iodine-refractory differentiated thyroid cancer following failure with sorafenib or lenvatinib adds to the available armamentarium of active agents. Vandetanib and cabozantinib have become mainstay treatments for metastatic medullary thyroid cancer regardless of RET mutation status. Selpercatinib and pralsetinib, potent and selective receptor kinase inhibitors with activity against RET, have revolutionized the treatment paradigm for medullary thyroid cancers and other cancers with driver mutations in RET . Dabrafenib plus trametinib for BRAF mutated anaplastic thyroid cancer provides an effective treatment option for this aggressive cancer with a dismal prognosis. In order to design the next generation of agents for thyroid cancer, future efforts will need to focus on developing a better understanding of the mechanisms of resistance to kinase inhibition including bypass signaling and escape mutations., Competing Interests: MS declares research funding from Merck and Eli-Lilly. BK declares research funding (to institution) from Eli Lilly & Co, Merck, Eisai, and Xencor. VS declares research funding from Eli Lilly & Co., (© the author(s).)
- Published
- 2023
- Full Text
- View/download PDF
35. Culture impact on the transcriptomic programs of primary and iPSC-derived human alveolar type 2 cells.
- Author
-
Alysandratos KD, Garcia-de-Alba C, Yao C, Pessina P, Huang J, Villacorta-Martin C, Hix OT, Minakin K, Burgess CL, Bawa P, Murthy A, Konda B, Beers MF, Stripp BR, Kim CF, and Kotton DN
- Subjects
- Humans, Animals, Mice, Transcriptome, Alveolar Epithelial Cells metabolism, Lung pathology, Pulmonary Alveoli pathology, Induced Pluripotent Stem Cells
- Abstract
Dysfunction of alveolar epithelial type 2 cells (AEC2s), the facultative progenitors of lung alveoli, is implicated in pulmonary disease pathogenesis, highlighting the importance of human in vitro models. However, AEC2-like cells in culture have yet to be directly compared to their in vivo counterparts at single-cell resolution. Here, we performed head-to-head comparisons among the transcriptomes of primary (1°) adult human AEC2s, their cultured progeny, and human induced pluripotent stem cell-derived AEC2s (iAEC2s). We found each population occupied a distinct transcriptomic space with cultured AEC2s (1° and iAEC2s) exhibiting similarities to and differences from freshly purified 1° cells. Across each cell type, we found an inverse relationship between proliferative and maturation states, with preculture 1° AEC2s being most quiescent/mature and iAEC2s being most proliferative/least mature. Cultures of either type of human AEC2s did not generate detectable alveolar type 1 cells in these defined conditions; however, a subset of iAEC2s cocultured with fibroblasts acquired a transitional cell state described in mice and humans to arise during fibrosis or following injury. Hence, we provide direct comparisons of the transcriptomic programs of 1° and engineered AEC2s, 2 in vitro models that can be harnessed to study human lung health and disease.
- Published
- 2023
- Full Text
- View/download PDF
36. Cryobanking of Human Distal Lung Epithelial Cells for Preservation of Their Phenotypic and Functional Characteristics.
- Author
-
Konda B, Mulay A, Yao C, Israely E, Beil S, Huynh CA, Tourtellotte WG, Rampolla R, Chen P, Carraro G, and Stripp BR
- Subjects
- Humans, Mice, Animals, Cell Differentiation physiology, Alveolar Epithelial Cells metabolism, Phenotype, Lung, Epithelial Cells metabolism
- Abstract
The epithelium lining airspaces of the human lung is maintained by regional stem cells, including basal cells of pseudostratified airways and alveolar type 2 (AT2) pneumocytes of the gas-exchange region. Despite effective techniques for long-term preservation of airway basal cells, procedures for efficient preservation of functional epithelial cell types of the distal gas-exchange region are lacking. Here we detail a method for cryobanking of epithelial cells from either mouse or human lung tissue for preservation of their phenotypic and functional characteristics. Flow cytometric profiling, epithelial organoid-forming efficiency, and single-cell transcriptomic analysis were used to compare cells recovered from cryobanked tissue with those of freshly dissociated tissue. AT2 cells within single-cell suspensions of enzymatically digested cryobanked distal lung tissue retained expression of the pan-epithelial marker CD326 and the AT2 cell surface antigen recognized by monoclonal antibody HT II-280, allowing antibody-mediated enrichment and downstream analysis. Isolated AT2 cells from cryobanked tissue were comparable with those of freshly dissociated tissue both in their single-cell transcriptome and their capacity for in vitro organoid formation in three-dimensional cultures. We conclude that the cryobanking method described herein allows long-term preservation of distal human lung tissue for downstream analysis of lung cell function and molecular phenotype and is ideally suited for the creation of an easily accessible tissue resource for the research community.
- Published
- 2022
- Full Text
- View/download PDF
37. Dabrafenib Versus Dabrafenib + Trametinib in BRAF -Mutated Radioactive Iodine Refractory Differentiated Thyroid Cancer: Results of a Randomized, Phase 2, Open-Label Multicenter Trial.
- Author
-
Busaidy NL, Konda B, Wei L, Wirth LJ, Devine C, Daniels GA, DeSouza JA, Poi M, Seligson ND, Cabanillas ME, Sipos JA, Ringel MD, Eisfeld AK, Timmers C, and Shah MH
- Subjects
- Humans, Adolescent, Adult, Proto-Oncogene Proteins B-raf genetics, Iodine Radioisotopes therapeutic use, Pyrimidinones adverse effects, Pyridones adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Oximes adverse effects, Mutation, Melanoma drug therapy, Thyroid Neoplasms drug therapy, Thyroid Neoplasms genetics, Thyroid Neoplasms radiotherapy, Adenocarcinoma
- Abstract
Background: Oncogenic BRAF mutations are commonly found in advanced differentiated thyroid cancer (DTC), and reports have shown efficacy of BRAF inhibitors in these tumors. We investigated the difference in response between dabrafenib monotherapy and dabrafenib + trametinib therapy in patients with BRAF-mutated radioactive iodine refractory DTC. Methods: In this open-label randomized phase 2 multicenter trial, patients aged ≥18 years with BRAF-mutated radioactive iodine refractory DTC with progressive disease by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 within 13 months before enrollment were eligible. Patients were randomly assigned to receive dabrafenib alone or dabrafenib + trametinib. The primary endpoint was objective response rate by modified RECIST (minor response of -20% to -29%, partial and complete response) within the first 24 weeks of therapy. Trial Registration Number: NCT01723202. Results: A total of 53 patients were enrolled. The objective response rate (modified RECIST) was 42% (11/26 [95% confidence interval {CI} 23-63%]) with dabrafenib versus 48% (13/27 [CI 29-68%]) with dabrafenib + trametinib ( p = 0.67). Objective response rate (RECIST 1.1) was 35% (9/26 [CI 17-56%]) with dabrafenib and 30% (8/27 [CI 14-51%]) with dabrafenib + trametinib. Most common treatment-related adverse events included skin and subcutaneous tissue disorders (17/26, 65%), fever (13/26, 50%), hyperglycemia (12/26, 46%) with dabrafenib alone and fever (16/27, 59%), nausea, chills, fatigue (14/27, 52% each) with dabrafenib + trametinib. There were no treatment-related deaths. Conclusions: Combination dabrafenib + trametinib was not superior in efficacy compared to dabrafenib monotherapy in patients with BRAF-mutated radioiodine refractory progressive DTC.
- Published
- 2022
- Full Text
- View/download PDF
38. Tumour-agnostic efficacy and safety of selpercatinib in patients with RET fusion-positive solid tumours other than lung or thyroid tumours (LIBRETTO-001): a phase 1/2, open-label, basket trial.
- Author
-
Subbiah V, Wolf J, Konda B, Kang H, Spira A, Weiss J, Takeda M, Ohe Y, Khan S, Ohashi K, Soldatenkova V, Szymczak S, Sullivan L, Wright J, and Drilon A
- Subjects
- Alanine Transaminase, Aspartate Aminotransferases, Humans, Protein Kinase Inhibitors adverse effects, Proto-Oncogene Proteins c-ret genetics, Pyrazoles, Pyridines, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Thyroid Neoplasms drug therapy, Thyroid Neoplasms genetics
- Abstract
Background: Selpercatinib is a first-in-class, highly selective RET kinase inhibitor with CNS activity that has shown efficacy in RET fusion-positive lung and thyroid cancers. RET fusions occur rarely in other tumour types. We aimed to investigate the efficacy and safety of selpercatinib in a diverse group of patients with RET fusion-positive non-lung or thyroid advanced solid tumours (ie, a tumour-agnostic population)., Methods: LIBRETTO-001 is an ongoing phase 1/2, single-group, open-label, basket trial of selpercatinib in patients aged 18 years and older (or ≥12 years, where permitted by regulatory authorities) with RET-altered cancers. The trial is being conducted at 89 sites in 16 countries; the tumour-agnostic population was enrolled at 30 sites (outpatient and inpatient medical facilities) across eight countries. A prespecified interim analysis of LIBRETTO-001 was planned to investigate the efficacy and safety of selpercatinib in a tumour-agnostic population of patients with RET fusion-positive advanced solid tumours; the data cutoff date was Sept 24, 2021. Eligible patients had disease progression on or after previous systemic therapies or no satisfactory therapeutic options and an Eastern Cooperative Oncology Group performance status of 0-2. Selpercatinib was orally administered in a continuous 28-day cycle. Patients enrolled in the phase 1 dose-escalation portion received between 20 mg once daily or 20-240 mg twice daily; the phase 2 recommended dose was 160 mg twice daily. The primary endpoint was the objective response rate as determined by the independent review committee. The efficacy-evaluable tumour-agnostic population was defined as patients with RET fusion-positive cancer, other than non-small-cell lung cancer and thyroid cancer, who had at least 6 months of follow-up from the first study dose at the time of data cutoff (all responders at the time of data cutoff were followed up for at least 6 months from the onset of response unless they progressed or died earlier). Safety was analysed in the tumour-agnostic population of patients who had been enrolled and received selpercatinib on or before the data cutoff date. This study is registered with ClinicalTrials.gov (NCT03157128) and is still recruiting participants., Findings: Between Dec 4, 2017, and Aug 4, 2021, 45 patients with RET fusion-positive tumour-agnostic cancers were enrolled from the phase 1 dose-escalation and phase 2 dose-expansion cohorts of the trial. 43 (96%) of 45 patients received a starting dose of selpercatinib at the recommended dose of 160 mg twice daily. Of the two patients who did not, one received a dose of 160 mg twice daily via intra-patient dose escalation (as allowed per protocol for patients enrolled in the phase 1 portion of the study at lower doses) and the other patient's starting dose of 120 mg twice daily was never escalated. Of the 41 efficacy-evaluable patients, the objective response rate as per the independent review committee was 43·9% (95% CI 28·5-60·3; 18 of 41 patients). The most common grade 3 or worse treatment-emergent adverse events were hypertension (ten [22%] of 45 patients), increased alanine aminotransferase (seven [16%]), and increased aspartate aminotransferase (six [13%]). Treatment-emergent serious adverse events occurred in 18 (40%) of 45 patients. No treatment-related deaths occurred., Interpretation: Selpercatinib showed clinically meaningful activity in the RET fusion-positive tumour-agnostic population, with a safety profile consistent with that observed in other indications. Comprehensive genomic testing that includes RET fusions will be crucial for identifying patients who might benefit from selpercatinib., Funding: Loxo Oncology., Competing Interests: Declaration of interests VSu reports grants from Blueprint Medicines, Boston Pharmaceuticals, Eli Lilly/Loxo Oncology, and Turning Point Therapeutics; grants from Helsinn Pharmaceuticals during the conduct of the study; a grant and advisory board or consultant position with Eli Lilly/Loxo Oncology during the conduct of the study; research grants from AbbVie, Agensys, Alfa-sigma, Altum, Amgen, Bayer, Berghealth, Blueprint Medicines, Boston Biomedical, Boston Pharmaceuticals, Celgene, D3, Dragonfly Therapeutics, Exelixis, Fujifilm, GlaxoSmithKline, Idera Pharma, Incyte, Inhibrx, MedImmune, Multivir, Nanocarrier, National Comprehensive Cancer Network, NCI-CTEP, Northwest Biotherapeutics, Novartis, Pfizer, Pharmamar, Roche/Genentech, Takeda, Turning Point Therapeutics, University of Texas MD Anderson Cancer Center, and Vegenics; advisory board and consultant positions with Daiichi Sankyo, Helsinn, Incyte, MedImmune, Novartis, QED Pharma, Relay Therapeutics, Roche, and Signant Health; travel funds from ASCO, ESMO, Incyte, and Pharmamar; educational grant support from Medscape; all outside the submitted work. JWo reports personal grants from Amgen, AstraZeneca, Bayer, Blueprint Medicines, Bristol Myers Squibb, Boehringer Ingelheim, Chugai, Daiichi Sankyo, Eli Lilly and Company, Ignyta, Janssen, Loxo Oncology, MSD, Novartis, Pfizer, Roche, Seattle Genetics, and Takeda; and institutional grants from Bristol Myers Squibb, Janssen, Novartis, and Pfizer. BK reports institutional grants from Bristol Myers Squibb, Eisai, Eli Lilly and Company, Merck, and Xencor. AS reports personal funding support from Loxo Oncology/Eli Lilly and Company; institutional grants from AbbVie, ADCT, Amgen, Arch Therapeutics, Astellas Pharma, Astex Pharmaceuticals, AstraZeneca, Blueprint Medicines, Boehringer Ingelheim, Bristol Myers Squibb, CytomX Therapeutics, Daiichi Sankyo, Gritstone, Ignyta, Incyte, Janssen Oncology, LAM Therapeutics, Loxo Oncology, Macrogenics, MedImmune, Mersana, Mirati Therapeutics, Newlink Genetics, Novartis, Plexxikon, Roche, Rubius, Synthekine, Takeda, and Trovagene; consulting fees from Amgen, Array BioPharma, AstraZeneca/MedImmune, Blueprint Medicines, Bristol Myers Squibb, Daiichi Sankyo/AstraZeneca, Gritstone Bio, Gritstone Oncology, Incyte, Janssen, Jazz Pharmaceuticals, Merck, Mersana, Mirati Therapeutics, Novartis, and Takeda; honoraria from Amgen, AstraZeneca/MedImmune, Bayer, Bristol Myers Squibb, CytomX Therapeutics, Janssen Oncology, Merck, Novartis, and Takeda; and stock options from Eli Lilly and Company. JWe reports support for the present manuscript from Eli Lilly and Company; grants from Amgen, AstraZeneca, Boehringer Ingelheim, Carefusion, G1, Immunicum, Merck, Mirati, PDS, and SDP; consulting fees from AbbVie, AstraZeneca, Azitra, Boehringer Ingelheim, Blueprint Medicines, Genentech, Genmab, G1, Jazz, Nanobiotix, Pfizer, Regeneron, Saatchi, SDP, and Wellness; travel support from Mirati; advisory board participation for EMD Serono and Jounce; and stock options with En Fuego Therapeutics (convertible note), Iovance, Lyel, Nektar, and Nuvalen. MT reports honoraria from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Chugai, Eli Lilly and Company, Novartis Pharma, ONO Pharmaceutical, and Takeda Pharma. YO reports grants from AstraZeneca, Bristol Myers Squibb, Chugai, Daiichi Sankyo, Dainippon-Sumitomo, Eli Lilly and Company, Janssen, Kissei, Kyorin, Loxo Oncology, Novartis, ONO, Pfizer, Taiho, and Takeda; honoraria from AstraZeneca, Bayer, Bristol Myers Squibb, Boehringer Ingelheim, Chugai, Eli Lilly and Company, Kayaku, Kyowa Hakko Kirin, MSD, Nippon, ONO, Pfizer, and Taiho; expert testimony for Amgen, AnHeart Therapeutics, AstraZeneca, Bristol Myers Squibb, Boehringer Ingelheim, Celltrion, Chugai, Kayaku, Kyorin, Nippon, and ONO; and leadership roles with the Japan Clinical Oncology Group, Japan Lung Cancer Society, and Japanese Society of Medical Oncology. SK reports grants from AbbVie, Bayer/Onyx, Bristol Myers Squibb, Celldex, Formation Biologics, Genentech, Gilead Sciences, Guardant Health, Loxo Oncology, Merck, Novartis, Pfizer, and Threshold Pharmaceuticals; and consulting fees from Eisai, Foundation Medicine, and Genentech/Roche. VSo, SS, LS, and JWr report employment and stock options with Eli Lilly and Company. AD reports consulting fees from AbbVie, Applied Pharmaceutical, ArcherDx, AstraZeneca, AXIS, BergenBio, Blueprint Medicines, Clinical Care Options, Entos, EPG Health, Genentech, Hengrui Therapeutics, i3 Health, Ignyta, the Journal of the National Comprehensive Cancer Network, Liberum, Loxo Oncology, Bayer, Eli Lilly and Company, mBrace, Med Learning, Medscape, Merus, MORE Health, Nuvalent, Ology, PeerView, Pfizer, Prelude, Roche, TouchIME, Treeline Bio, and Tyra Biosciences; and honoraria from 14ner/Elevation Oncology, AbbVie, AiCME, Amgen, Applied Pharmaceutical, Archer DX, Ariad, AstraZeneca, AXIS, BeiGene, BergenBio, Blueprint Medicines, Chugai, EMD Serono, Entos, EPG Health, Exelixis, Genentech, Harborside Nexus, Helsinn, Hengrui Therapeutics, i3 Health, Ignyta, Janssen, Liberum, Loxo Oncology, Bayer, Eli Lilly and Company, mBrace, Medendi, Merus, Millenium, Monopteros, MonteRosa, MORE Health, Novartis, Nuvalent, Ology, Pfizer, Prelude, Remedica, Repare Rx, Roche, RV More, Takeda, TouchIME, TP Therapeutics, Treeline Bio, Tyra Biosciences, and Verastem; advisory board participation for 14ner/Elevation Oncology, Amgen, Janssen, Loxo Oncology, Bayer, Eli Lilly and Company, Melendi, MonteRosa, Novartis, Pfizer, and Repare Rx; associated research to their institution from Exelixis, GlaxoSmithKlein, Pfizer, PharmaMar, Taiho, and Teva; royalties from Wolters Kluwer; a patent for selpercatinib and osimertinib (pending; WO 2022/046867); stock options from Treeline Bio; and other support (food or beverage) from Boehringer Ingelheim, Merck, Merus, and Puma. HK and KO declare no competing interests., (Copyright © 2022 Elsevier Ltd. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
39. Pulmonary infection by SARS-CoV-2 induces senescence accompanied by an inflammatory phenotype in severe COVID-19: possible implications for viral mutagenesis.
- Author
-
Evangelou K, Veroutis D, Paschalaki K, Foukas PG, Lagopati N, Dimitriou M, Papaspyropoulos A, Konda B, Hazapis O, Polyzou A, Havaki S, Kotsinas A, Kittas C, Tzioufas AG, de Leval L, Vassilakos D, Tsiodras S, Stripp BR, Papantonis A, Blandino G, Karakasiliotis I, Barnes PJ, and Gorgoulis VG
- Subjects
- Cellular Senescence, Cytokines metabolism, Humans, Inflammation, Interleukin-6, Lung metabolism, Mutagenesis, Phenotype, COVID-19, SARS-CoV-2
- Abstract
Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of the respiratory system can progress to a multisystemic disease with aberrant inflammatory response. Cellular senescence promotes chronic inflammation, named senescence-associated secretory phenotype (SASP). We investigated whether coronavirus disease 2019 (COVID-19) is associated with cellular senescence and SASP., Methods: Autopsy lung tissue samples from 11 COVID-19 patients and 43 age-matched non-COVID-19 controls with similar comorbidities were analysed by immunohistochemistry for SARS-CoV-2, markers of senescence and key SASP cytokines. Virally induced senescence was functionally recapitulated in vitro , by infecting epithelial Vero-E6 cells and a three-dimensional alveosphere system of alveolar type 2 (AT2) cells with SARS-CoV-2 strains isolated from COVID-19 patients., Results: SARS-CoV-2 was detected by immunocytochemistry and electron microscopy predominantly in AT2 cells. Infected AT2 cells expressed angiotensin-converting enzyme 2 and exhibited increased senescence (p16
INK4A and SenTraGor positivity) and interleukin (IL)-1β and IL-6 expression. In vitro , infection of Vero-E6 cells with SARS-CoV-2 induced senescence (SenTraGor), DNA damage (γ-H2AX) and increased cytokine (IL-1β, IL-6, CXCL8) and apolipoprotein B mRNA-editing (APOBEC) enzyme expression. Next-generation sequencing analysis of progenies obtained from infected/senescent Vero-E6 cells demonstrated APOBEC-mediated SARS-CoV-2 mutations. Dissemination of the SARS-CoV-2-infection and senescence was confirmed in extrapulmonary sites (kidney and liver) of a COVID-19 patient., Conclusions: We demonstrate that in severe COVID-19, AT2 cells infected by SARS-CoV-2 exhibit senescence and a proinflammatory phenotype. In vitro , SARS-CoV-2 infection induces senescence and inflammation. Importantly, infected senescent cells may act as a source of SARS-CoV-2 mutagenesis mediated by APOBEC enzymes. Therefore, SARS-CoV-2-induced senescence may be an important molecular mechanism of severe COVID-19, disease persistence and mutagenesis., Competing Interests: Conflict of interest: P.J. Barnes receives research funding from AstraZeneca and Boehringer Ingelheim and is a scientific advisor to AstraZeneca, Boehringer Ingelheim, Epi-Endo, Novartis, Pieris and Teva. The other authors wish to declare no conflict of interest., (Copyright ©The authors 2022.)- Published
- 2022
- Full Text
- View/download PDF
40. An Evaluation of Clinical Efficacy of Immune Checkpoint Inhibitors for Patients with Anaplastic Thyroid Carcinoma.
- Author
-
Hatashima A, Archambeau B, Armbruster H, Xu M, Shah M, Konda B, Lott Limbach A, and Sukrithan V
- Subjects
- Aged, B7-H1 Antigen, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Male, Treatment Outcome, Thyroid Carcinoma, Anaplastic pathology, Thyroid Neoplasms pathology
- Abstract
Background: Anaplastic thyroid carcinoma (ATC) is an aggressive thyroid malignancy that is associated with poor prognosis. Current treatment options include surgery, radiation, cytotoxic chemotherapy, and multikinase inhibitor therapy. The role of immunotherapy in ATC is an area of active interest and recent evidence suggests that it may be a potentially effective treatment option. Methods: We report a case series of 13 patients with locally advanced or metastatic unresectable ATC who received immune checkpoint inhibitor therapy (pembrolizumab or nivolumab) at a single institution. Results: The patients' median age was 70 years, 54% (7/13) were male, and 85% (11/13) had stage IVC disease with lungs and lymph nodes being the most common sites of metastases. Ten patients had tumor tissue available for programmed death-ligand 1 (PD-L1) expression testing, all of which were positive for PD-L1, and seven of these patients also had a BRAF
V600E mutation. The median progression-free survival was 1.9 months and median overall survival (OS) was 4.4 months. The objective response rate was 16% (2/13). Two patients had partial response (PR), and three patients had durable stable disease. Among patients with a clinical benefit, after a median follow-up of 13.5 months, median OS had not been reached (range 4+ to 29+ months). Responses were ongoing in four subjects. The one-year survival rate was 38% (5/13). Six patients (46%) experienced an immune-related adverse event, and 15% (2/13) experienced a grade 3 or higher adverse event, including one patient with grade 5 immune checkpoint-related thyroiditis. Conclusions: Immune checkpoint blockade was well tolerated with a toxicity profile consistent with published literature on PD-1/PD-L1-targeting therapies. For ATC patients, immune checkpoint inhibition may represent an effective treatment option with robust sustained responses seen in a subset of patients.- Published
- 2022
- Full Text
- View/download PDF
41. A Randomized Study of Lenvatinib 18 mg vs 24 mg in Patients With Radioiodine-Refractory Differentiated Thyroid Cancer.
- Author
-
Brose MS, Panaseykin Y, Konda B, de la Fouchardiere C, Hughes BGM, Gianoukakis AG, Joo Park Y, Romanov I, Krzyzanowska MK, Leboulleux S, Binder TA, Dutcus C, Xie R, and Taylor MH
- Subjects
- Adult, Aged, Aged, 80 and over, Chemoradiotherapy adverse effects, Dose-Response Relationship, Drug, Double-Blind Method, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Phenylurea Compounds adverse effects, Progression-Free Survival, Protein Kinase Inhibitors adverse effects, Quinolines adverse effects, Radiation Tolerance, Response Evaluation Criteria in Solid Tumors, Thyroid Neoplasms mortality, Thyroid Neoplasms pathology, Young Adult, Chemoradiotherapy methods, Iodine Radioisotopes therapeutic use, Phenylurea Compounds administration & dosage, Protein Kinase Inhibitors administration & dosage, Quinolines administration & dosage, Thyroid Neoplasms therapy
- Abstract
Background: Lenvatinib is a multikinase inhibitor approved to treat radioiodine-refractory differentiated thyroid cancer (RR-DTC) at a starting dose of 24 mg/day. This study explored, in a double-blinded fashion, whether a starting dose of 18 mg/day would provide comparable efficacy with reduced toxicity., Methods: Patients with RR-DTC were randomized to lenvatinib 24 mg/day or 18 mg/day. The primary efficacy endpoint was objective response rate as of week 24 (ORRwk24); the odds ratio noninferiority margin was 0.4. The primary safety endpoint was frequency of grade ≥3 treatment-emergent adverse events (TEAEs) as of week 24. Tumors were assessed using RECIST v1.1. TEAEs were monitored and recorded., Results: The ORRwk24 was 57.3% (95% CI 46.1, 68.5) in the lenvatinib 24-mg arm and 40.3% (95% CI 29.3, 51.2) in the lenvatinib 18-mg arm, with an odds ratio (18/24 mg) of 0.50 (95% CI 0.26, 0.96). As of week 24, the rates of TEAEs grade ≥3 were 61.3% in the lenvatinib 24-mg arm and 57.1% in the lenvatinib 18-mg arm, a difference of -4.2% (95% CI -19.8, 11.4)., Conclusion: A starting dose of lenvatinib 18 mg/day did not demonstrate noninferiority compared to a starting dose of 24 mg/day as assessed by ORRwk24 in patients with RR-DTC. The results represent a clinically meaningful difference in ORRwk24. The safety profile was comparable, with no clinically relevant difference between arms. These results support the continued use of the approved starting dose of lenvatinib 24 mg/day in patients with RR-DTC and adjusting the dose as necessary., (© The Author(s) 2021. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.