75 results on '"Hegde PS"'
Search Results
2. Blood-Based Biomarkers for Cancer Immunotherapy: Tumor Mutational Burden in Blood (bTMB) is Associated with Improved Atezolizumab (atezo) Efficacy in 2L+ NSCLC (POPLAR and OAK)
- Author
-
Rittmeyer, A, additional, Gandara, D, additional, Kowanetz, M, additional, Mok, T, additional, Fehrenbacher, L, additional, Fabrizio, D, additional, Otto, G, additional, Malboeuf, C, additional, Lieber, D, additional, Paul, SM, additional, Amler, L, additional, Riehl, T, additional, Schleifman, E, additional, Cummings, C, additional, Hegde, PS, additional, Zou, W, additional, Sandler, A, additional, Ballinger, M, additional, and Shames, DS, additional
- Published
- 2018
- Full Text
- View/download PDF
3. Clinical Efficacy of atezolizumab (atezo) in PD-L1 subgroups defined by SP142 and 22C3 IHC assays in 2L+ NSCLC: Results from the randomized OAK study
- Author
-
Rittmeyer, A, additional, Gadgeel, S, additional, Kowanetz, M, additional, Zou, W, additional, Hirsch, FR, additional, Kerr, KM, additional, Gandara, D, additional, Barlesi, F, additional, Park, K, additional, McCleland, M, additional, Koeppen, H, additional, Ballinger, M, additional, Sandler, A, additional, and Hegde, PS, additional
- Published
- 2018
- Full Text
- View/download PDF
4. Novel technologies and emerging biomarkers for personalized cancer immunotherapy
- Author
-
Yuan, J, Hegde, PS, Clynes, R, Foukas, PG, Harari, A, Kleen, TO, Kvistborg, P, Maccalli, C, Maecker, HT, Page, DB, Robins, H, Song, W, Stack, EC, Wang, E, Whiteside, TL, Zhao, Y, Zwierzina, H, Butterfield, LH, Fox, BA, Yuan, J, Hegde, PS, Clynes, R, Foukas, PG, Harari, A, Kleen, TO, Kvistborg, P, Maccalli, C, Maecker, HT, Page, DB, Robins, H, Song, W, Stack, EC, Wang, E, Whiteside, TL, Zhao, Y, Zwierzina, H, Butterfield, LH, and Fox, BA
- Abstract
The culmination of over a century's work to understand the role of the immune system in tumor control has led to the recent advances in cancer immunotherapies that have resulted in durable clinical responses in patients with a variety of malignancies. Cancer immunotherapies are rapidly changing traditional treatment paradigms and expanding the therapeutic landscape for cancer patients. However, despite the current success of these therapies, not all patients respond to immunotherapy and even those that do often experience toxicities. Thus, there is a growing need to identify predictive and prognostic biomarkers that enhance our understanding of the mechanisms underlying the complex interactions between the immune system and cancer. Therefore, the Society for Immunotherapy of Cancer (SITC) reconvened an Immune Biomarkers Task Force to review state of the art technologies, identify current hurdlers, and make recommendations for the field. As a product of this task force, Working Group 2 (WG2), consisting of international experts from academia and industry, assembled to identify and discuss promising technologies for biomarker discovery and validation. Thus, this WG2 consensus paper will focus on the current status of emerging biomarkers for immune checkpoint blockade therapy and discuss novel technologies as well as high dimensional data analysis platforms that will be pivotal for future biomarker research. In addition, this paper will include a brief overview of the current challenges with recommendations for future biomarker discovery.
- Published
- 2016
5. Education reduces unnecessary diagnostic blood sampling in the intensive care unit (ICU)
- Author
-
Hegde, PS, Tarsey, K, and Blunt, MC
- Subjects
Meeting Abstract - Published
- 2003
6. A modification of the blood sampling technique in critical care to reduce blood wastage
- Author
-
Hegde, PS, Tarsey, K, and Blunt, MC
- Subjects
Meeting Abstract - Published
- 2003
7. Atezolizumab in combination with bevacizumab enhances migration of antigen-specific T-cells in metastatic renal cell carcinoma
- Author
-
Wallin, JJ, primary, Bendell, JC, additional, Funke, R, additional, Sznol, M, additional, Korski, K, additional, Jones, S, additional, Hernandez, G, additional, Mier, J, additional, He, X, additional, Hodi, FS, additional, Denker, M, additional, Leveque, V, additional, Cañamero, M, additional, Babitski, G, additional, Koeppen, H, additional, Aiai, J, additional, Sharma, N, additional, Gaire, F, additional, Chen, DS, additional, Waterkamp, D, additional, Hegde, PS, additional, and McDermott, DF, additional
- Full Text
- View/download PDF
8. Measurable residual mutated IDH1 before allogeneic transplant for acute myeloid leukemia.
- Author
-
Gui G, Ravindra N, Hegde PS, Andrew G, Mukherjee D, Wong Z, Auletta JJ, El Chaer F, Chen EC, Chen YB, Corner A, Devine SM, Iyer SG, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Saber W, Spellman SR, Zeger SL, Page KM, Dillon LW, and Hourigan CS
- Abstract
Measurable residual disease (MRD) in adults with acute myeloid leukemia (AML) in complete remission is an important prognostic marker, but detection methodology requires optimization. Persistence of mutated NPM1 or FLT3-ITD in the blood of adult patients with AML in first complete remission (CR1) prior to allogeneic hematopoietic cell transplant (alloHCT) associates with increased relapse and death after transplant. The prognostic implications of persistence of other common AML-associated mutations, such as IDH1, at this treatment landmark however remain incompletely defined. We performed testing for residual IDH1 variants (IDH1m) in pre-transplant CR1 blood of 148 adult patients undergoing alloHCT for IDH1-mutated AML at a CIBMTR reporting site between 2013 and 2019. No statistically significant post-transplant differences were observed between those testing IDH1m positive (n = 53, 36%) and negative pre-transplant (overall survival (OS): p = 0.4; relapse: p = 0.5). For patients with IDH1 mutated AML co-mutated with NPM1 and/or FLT3-ITD, only detection of persistent mutated NPM1 and/or FLT3-ITD was associated with significantly higher rates of relapse (p = 0.01). These data, from the largest study to date, do not support the detection of IDH1 mutation in CR1 blood prior to alloHCT as evidence of AML MRD for increased post-transplant relapse risk., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
9. Astaxanthin and DHA Supplementation Modulates the Maternal Undernutrition-induced Impairment of Cognitive Behavior and Synaptic Plasticity in Adult Life of Offspring's -Exploring the Molecular Mechanism.
- Author
-
Agni MB, Hegde PS, Rai P, Sadananda M, and K M DG
- Subjects
- Animals, Female, Pregnancy, Rats, Wistar, Maternal Nutritional Physiological Phenomena, Male, Cognition drug effects, Hippocampus metabolism, Hippocampus drug effects, Animals, Newborn, Prenatal Exposure Delayed Effects, Behavior, Animal drug effects, Neuronal Plasticity drug effects, Docosahexaenoic Acids pharmacology, Docosahexaenoic Acids therapeutic use, Dietary Supplements, Malnutrition complications, Xanthophylls pharmacology
- Abstract
Maternal nutrition was recognized as a significant part of brain growth and maturation in most mammalian species. Timely intervention with suitable nutraceuticals would provide long-term health benefits. We aim to unravel the molecular mechanisms of perinatal undernutrition-induced impairments in cognition and synaptic plasticity, employing animal model based on dietary nutraceutical supplementation. We treated undernourished dams at their gestational, lactational, and at both the time point with Astaxanthin (AsX) and Docosahexaenoic acid (DHA), and their pups were used as experimental animals. We evaluated the cognitive function by subjecting the pups to behavioral tests in their adult life. In addition, we assessed the expression of genes in the hippocampus related to cognitive function and synaptic plasticity. Our results showed downregulation of Brain-derived neurotrophic factor (BDNF), Neurotrophin-3 (NT-3), cAMP response-element-binding protein (CREB), and uncoupling protein-2 (UCP2) gene expression in pups born to undernourished dams in their adult life, which AsX and DHA modulated. Maternal AsX and DHA supplementation ameliorated the undernutrition-induced learning impairment in novel object recognition (NOR) tests and partially baited radial arm maze (RAM) tasks in offspring's. The expressions of Synapsin-1 and PSD-95 decreased in perinatally undernourished groups compared to control and AsX-DHA treated groups at CA1, CA2, CA3, and DG. AsX and DHA supplementation upregulated BDNF, NT-3, CREB, and UCP2 gene expressions in perinatally undernourished rats, which are involved in intracellular signaling cascades like Ras, PI3K, and PLC. The results of our study give new insights into neuronal differentiation, survival, and plasticity, indicating that the perinatal period is the critical time for reversing maternal undernutrition-induced cognitive impairment in offspring's., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
10. Measurable residual FLT3 tyrosine kinase domain mutations before allogeneic transplant for acute myeloid leukemia.
- Author
-
Hegde PS, Andrew G, Gui G, Ravindra N, Mukherjee D, Wong ZC, Auletta JJ, El Chaer F, Corner A, Devine SM, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Saber W, Spellman SR, Zeger SL, Page KM, Dillon LW, and Hourigan CS
- Published
- 2024
- Full Text
- View/download PDF
11. Analytical validation of a novel comprehensive genomic profiling informed circulating tumor DNA monitoring assay for solid tumors.
- Author
-
Zollinger DR, Rivers E, Fine A, Huang Y, Son J, Kalyan A, Gray W, Baharian G, Hammond C, Ram R, Ringman L, Hafez D, Savel D, Patel V, Dantone M, Guo C, Childress M, Xu C, Johng D, Wallden B, Pokharel P, Camara W, Hegde PS, Hughes J, Carter C, Davarpanah N, Degaonkar V, Gupta P, Mariathasan S, Powles T, Ferree S, Dennis L, and Young A
- Subjects
- Humans, Genomics methods, Biomarkers, Tumor blood, Biomarkers, Tumor genetics, Sensitivity and Specificity, Algorithms, Multiplex Polymerase Chain Reaction methods, Liquid Biopsy methods, Circulating Tumor DNA blood, Circulating Tumor DNA genetics, Neoplasms genetics, Neoplasms blood, Neoplasms diagnosis
- Abstract
Emerging technologies focused on the detection and quantification of circulating tumor DNA (ctDNA) in blood show extensive potential for managing patient treatment decisions, informing risk of recurrence, and predicting response to therapy. Currently available tissue-informed approaches are often limited by the need for additional sequencing of normal tissue or peripheral mononuclear cells to identify non-tumor-derived alterations while tissue-naïve approaches are often limited in sensitivity. Here we present the analytical validation for a novel ctDNA monitoring assay, FoundationOne®Tracker. The assay utilizes somatic alterations from comprehensive genomic profiling (CGP) of tumor tissue. A novel algorithm identifies monitorable alterations with a high probability of being somatic and computationally filters non-tumor-derived alterations such as germline or clonal hematopoiesis variants without the need for sequencing of additional samples. Monitorable alterations identified from tissue CGP are then quantified in blood using a multiplex polymerase chain reaction assay based on the validated SignateraTM assay. The analytical specificity of the plasma workflow is shown to be 99.6% at the sample level. Analytical sensitivity is shown to be >97.3% at ≥5 mean tumor molecules per mL of plasma (MTM/mL) when tested with the most conservative configuration using only two monitorable alterations. The assay also demonstrates high analytical accuracy when compared to liquid biopsy-based CGP as well as high qualitative (measured 100% PPA) and quantitative precision (<11.2% coefficient of variation)., Competing Interests: I have read the journal’s policy and the authors of this manuscript have the following competing interests: Stock and Employment from Foundation Medicine, Inc: D.R.Z., A.F., Y.H., C.G., M.C., C.X., D.J., B.W., P.P., W.C., P.H., J.H., L.D., A.Y. Stock and Employment from Natera: E.R., J.S., A.K., W.G., G.B., C.H., R.R., L.R., D.H., D.S., V.P., M.D., S.F. Stock and Employment from F. Hoffmann-La Roche: C.C., N.D., V.D., P.G., S.M. Honararia- Astellas, Pfizer, Seagen, BMS, Roche, Astra-Zeneca, MSD, Natera, Merck Serono, Johnson and Johnson - T.P. This does not alter our adherence to PLOS ONE policies on sharing data and materials, (Copyright: © 2024 Zollinger 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
12. Nutritional efficacy of Astaxanthin in modulating orexin peptides and fatty acid level during adult life of rats exposed to perinatal undernutrition stress.
- Author
-
Bhat Agni M, Hegde PS, Ullal H, and Damodara Gowda KM
- Subjects
- Pregnancy, Female, Rats, Male, Animals, Orexins, Rats, Wistar, Fatty Acids analysis, Malnutrition, Fatty Acids, Omega-3
- Abstract
Perinatal undernutrition stress predisposes several disorders in adult life, which could be programed using nutraceuticals. However, the effect of perinatal undernutrition stress on orexin peptides, brain lipids, and its amelioration by a potent antioxidant (Astaxanthin) needs exploration. The present study focussed on the effect of perinatal undernutrition stress on brain fatty acid levels, Orexin peptides A and B, and its amelioration by Astaxanthin.Twenty-four male Wistar rats ( Rattus norvegicus ) were allocated to four groups (n = 6) as Normal, Perinatally Undernourished (UN), Astaxanthin treated (AsX, 12mg/kg), and perinatally Undernourished-but-Astaxanthin treated (UNA), and are allowed to grow for 1, 6 and 12 months. The fatty acid and orexin peptides A & B at different brain parts were measured and compared. Orexin peptides were assessed using an ELISA kit. Fatty acid levels were estimated using HP 5890 gas chromatograph. Data were analyzed by ANOVA followed by Tukey's posthoc test. P < 0.05 was considered significant.The hair cortisol, Orexin-A, and B were significantly increased ( p < 0.001) in the UN group compared to normal and were modulated significantly by AsX in the UNA group. Undernutrition stress during the perinatal period altered the lipid profile, Total SFA, Total MUFA, Total n-3 PUFA , Total n-6 PUFA , n-3: n-6 PUFA, which Astaxanthin effectively modulated at 6 and 12 months of postnatal life. There was no difference between DHA and AA ratio. These results indicate that nutritional enrichment with Astaxanthin during the perinatal period positively contributes to adult health. Further, the mechanism of regulation of brain chemistry by Astaxanthin is warranted.
- Published
- 2023
- Full Text
- View/download PDF
13. Pan-Cancer Analysis of Copy-Number Features Identifies Recurrent Signatures and a Homologous Recombination Deficiency Biomarker to Predict Poly (ADP-Ribose) Polymerase Inhibitor Response.
- Author
-
Moore JA, Chen KT, Madison R, Newberg JY, Fleischmann Z, Wang S, Sharaf R, Murugesan K, Fendler BJ, Hughes J, Schrock AB, Hegde PS, Oxnard GR, Fabrizio D, Frampton GM, Antonarakis ES, Sokol ES, and Jin DX
- Subjects
- Male, Female, Humans, Ribose therapeutic use, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Recombinational DNA Repair, Biomarkers, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Antineoplastic Agents therapeutic use
- Abstract
Purpose: Copy-number (CN) features reveal the molecular state of cancers and may have predictive and prognostic value in the treatment of cancer. We sought to apply published CN analysis methods to a large pan-cancer data set and characterize ubiquitous CN signatures across tumor types, including potential utility for treatment selection., Methods: We analyzed the landscape of CN features in 260,333 pan-cancer samples. We examined the association of 10 signatures with genomic alterations and clinical characteristics and trained a machine learning classifier using CN and insertion and deletion features to detect homologous recombination deficiency signature (HRDsig) positivity. Clinical outcomes were assessed using a real-world clinicogenomic database (CGDB) of comprehensive genomic profiling linked to deidentified, electronic health record-derived clinical data., Results: CN signatures were prevalent across cancer types and associated with diverse processes including focal tandem duplications, seismic amplifications, genome-wide loss of heterozygosity (gLOH), and HRD. Our novel HRDsig outperformed gLOH in predicting BRCAness and effectively distinguished biallelic BRCA and homologous recombination-repair wild-type (HRRwt) samples pan-tumor, demonstrating high sensitivity to detect biallelic BRCA in ovarian (93%) and other HRD-associated cancers (80%-87%). Pan-tumor prevalence of HRDsig was 6.4%. HRRwt cases represented a significant fraction of the HRDsig-positive cohort, likely reflecting a population with nongenomic mechanisms of HRD. In ovarian and prostate CGDBs, HRDsig identified more patients than gLOH and had predictive value for poly (ADP-ribose) polymerase inhibitor (PARPi) benefit., Conclusion: Tumor CN profiles are informative, revealing diverse processes active in cancer. We describe the landscape of 10 CN signatures in a large pan-cancer cohort, including two associated with HRD. We trained a machine learning-based HRDsig that robustly identified BRCAness and associated with biallelic BRCA pan-tumor, and was predictive of PARPi benefit in real-world ovarian and prostate data sets.
- Published
- 2023
- Full Text
- View/download PDF
14. Measurable Residual IDH1 before Allogeneic Transplant for Acute Myeloid Leukemia.
- Author
-
Gui G, Dillon LW, Ravindra N, Hegde PS, Andrew G, Mukherjee D, Wong Z, Auletta J, El Chaer F, Chen E, Chen YB, Corner A, Devine SM, Iyer S, Jimenez Jimenez AM, De Lima MJG, Litzow MR, Kebriaei P, Spellman SR, Zeger SL, Page KM, and Hourigan CS
- Abstract
Measurable residual disease (MRD) in adults with acute myeloid leukemia (AML) in complete remission is an important prognostic marker, but detection methodology requires optimization. The persistence of mutated NPM1 or FLT3 -ITD in the blood of adult patients with AML in first complete remission (CR1) prior to allogeneic hematopoetic cell transplant (alloHCT) has been established as associated with increased relapse and death after transplant. The prognostic implications of persistence of other common AML-associated mutations, such as IDH1 , at this treatment landmark however remains incompletely defined. We performed testing for residual IDH1 variants ( IDH1 m) in pre-transplant CR1 blood of 148 adult patients undergoing alloHCT for IDH1 -mutated AML at a CIBMTR site between 2013-2019. No post-transplant differences were observed between those testing IDH1 m positive (n=53, 36%) and negative pre-transplant (overall survival: p = 0.4; relapse: p = 0.5). For patients with IDH1 mutated AML co-mutated with NPM1 and/or FLT3 -ITD, only detection of persistent mutated NPM1 and/or FLT3 -ITD was associated with significantly higher rates of relapse (p = 0.01). These data, from the largest study to date, do not support the detection of IDH1 mutation in CR1 blood prior to alloHCT as evidence of AML MRD or increased post-transplant relapse risk., Competing Interests: Conflicts of Interest Statements Hourigan: The National Heart, Lung, and Blood Institute receives research funding for the laboratory of Dr. Hourigan from the Foundation of the NIH AML MRD Biomarkers Consortium. Auletta: Advisory Committee: AscellaHealth and Takeda El Chaer: Consultant: SPD Oncology, Amgen, Association of Community Cancer Centers; Clinical Trial Grant Support (PI) to the University of Virginia: Amgen, BMS, Celgene, SPD Oncology, Sanofi, Bristol Myers Squibb, FibroGen, PharmaEssentia, BioSight, MEI Pharma, Novartis, Arog pharmaceuticals; Travel grant: DAVA Oncology E Chen: Consultant: Rigel Pharmaceuticals and AbbVie Corner: Employment: Bio-Rad Laboratories Jimenez Jimenez: Funding: Abbvie De Lima: Advisory Board: Pfizer, Bristol Myers Squibb; Data Safety Monitoring Board: Novartis, Abbvie; Research Funding: Miltenyi Biotec Kebriaei: Consultant: Pfizer, Jazz Pharmaceuticals
- Published
- 2023
- Full Text
- View/download PDF
15. Molecular residual disease detection in resected, muscle-invasive urothelial cancer with a tissue-based comprehensive genomic profiling-informed personalized monitoring assay.
- Author
-
Powles T, Young A, Nimeiri H, Madison RW, Fine A, Zollinger DR, Huang Y, Xu C, Gjoerup OV, Aushev VN, Wu HT, Aleshin A, Carter C, Davarpanah N, Degaonkar V, Gupta P, Mariathasan S, Schleifman E, Assaf ZJ, Oxnard G, and Hegde PS
- Abstract
Introduction: Circulating tumor DNA (ctDNA) detection postoperatively may identify patients with urothelial cancer at a high risk of relapse. Pragmatic tools building off clinical tumor next-generation sequencing (NGS) platforms could have the potential to increase assay accessibility., Methods: We evaluated the widely available Foundation Medicine comprehensive genomic profiling (CGP) platform as a source of variants for tracking of ctDNA when analyzing residual samples from IMvigor010 (ClinicalTrials.gov identifier NCT02450331), a randomized adjuvant study comparing atezolizumab with observation after bladder cancer surgery. Current methods often involve germline sampling, which is not always feasible or practical. Rather than performing white blood cell sequencing to filter germline and clonal hematopoiesis (CH) variants, we applied a bioinformatic approach to select tumor (non-germline/CH) variants for molecular residual disease detection. Tissue-informed personalized multiplex polymerase chain reaction-NGS assay was used to detect ctDNA postsurgically (Natera)., Results: Across 396 analyzed patients, prevalence of potentially actionable alterations was comparable with the expected prevalence in advanced disease (13% FGFR2/3 , 20% PIK3CA , 13% ERBB2 , and 37% with elevated tumor mutational burden ≥10 mutations/megabase). In the observation arm, 66 of the 184 (36%) ctDNA-positive patients had shorter disease-free survival [DFS; hazard ratio (HR) = 5.77; 95% confidence interval (CI), 3.84-8.67; P < 0.0001] and overall survival (OS; HR = 5.81; 95% CI, 3.41-9.91; P < 0.0001) compared with ctDNA-negative patients. ctDNA-positive patients had improved DFS and OS with atezolizumab compared with those in observation (DFS HR = 0.56; 95% CI, 0.38-0.83; P = 0.003; OS HR = 0.66; 95% CI, 0.42-1.05). Clinical sensitivity and specificity for detection of postsurgical recurrence were 58% (60/103) and 93% (75/81), respectively., Conclusion: We present a personalized ctDNA monitoring assay utilizing tissue-based FoundationOne
® CDx CGP, which is a pragmatic and potentially clinically scalable method that can detect low levels of residual ctDNA in patients with resected, muscle-invasive bladder cancer without germline sampling., Competing Interests: TP received honoraria from advisory/consultancy roles with AstraZeneca, BMS, Exelixis, Incyte, Ipsen, Merck, MSD, Novartis, Pfizer, Seattle Genetics, Merck Serono EMD Serono, Astellas, Johnson & Johnson, Eisai, Mashup Ltd, and Roche; institutional research funding support from AstraZeneca, Roche, BMS, Exelixis, Ipsen, Merck, MSD, Novartis, Pfizer, Seattle Genetics, Merck Serono EMD Serono, Astellas, Eisai, and Johnson & Johnson; and travel, accommodation, and expenses support from Roche, Pfizer, MSD, AstraZeneca, and Ipsen. AY, HN, RM, AF, DZ, YH, CX, OG, GO, and PH are employees of Foundation Medicine, a wholly owned subsidiary of Roche, and have equity interest in Roche. VA, HW, and AA are employees of Natera, Inc., and reports stock ownership in Natera. CC, ND, VD, PG, SM, ES, and ZA are employees of Genentech and have equity interest in Roche., (Copyright © 2023 Powles, Young, Nimeiri, Madison, Fine, Zollinger, Huang, Xu, Gjoerup, Aushev, Wu, Aleshin, Carter, Davarpanah, Degaonkar, Gupta, Mariathasan, Schleifman, Assaf, Oxnard and Hegde.)- Published
- 2023
- Full Text
- View/download PDF
16. Astaxanthin and DHA supplementation ameliorates the proteomic profile of perinatal undernutrition-induced adipose tissue dysfunction in adult life.
- Author
-
Ranade AV, Hegde PS, Bhat MA, Rai P, Vinodini NA, Aravind A, Prasad TSK, and Damodara Gowda KM
- Subjects
- Female, Pregnancy, Animals, Rats, Chromatography, Liquid, Proteomics, Tandem Mass Spectrometry, Obesity, Adipose Tissue, Dietary Supplements, Docosahexaenoic Acids pharmacology, Malnutrition complications
- Abstract
Maternal diet is an essential factor that directly and indirectly regulates fetal growth. Exposure to certain environmental conditions substantially impacts an individual's short- and long-term health. Adipose tissue dysfunction is a worldwide chronic disease caused by improper lipid build-up in adipose tissue leading to obesity. Therefore, it is the need of the hour to invent anti-obesity agents. As a keto-carotenoid, Astaxanthin (AsX) has been shown to have preventive effects against problems associated with obesity. A crucial role in the pathogenesis of obesity has been attributed to dietary polyunsaturated fatty acids. Adipose tissue plays a vital role in maintaining overall body homeostasis. Metabolic dysfunction of white adipocytes forms a critical step in the emergence of insulin resistance and related diseases. Here we aim to investigate the effect of AsX and Docosahexaenoic acid (DHA) supplementation on the proteomic profile of perinatal undernutrition-induced adipose tissue dysfunction in adult life using a rat model. The LC-MS/MS quantitative proteomics enabled us to identify differentially expressed proteins in perinatal undernourished but AsX and DHA-supplemented animal models. Data are available via ProteomeXchange with identifier PXD041772.This study explored biological roles, molecular functions of differentially expressed proteins, and pathways related to adipose tissue dysfunction induced by undernutrition and its effective modulation by AsX and DHA., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
17. Integrative Analysis of a Large Real-World Cohort of Small Cell Lung Cancer Identifies Distinct Genetic Subtypes and Insights into Histologic Transformation.
- Author
-
Sivakumar S, Moore JA, Montesion M, Sharaf R, Lin DI, Colón CI, Fleishmann Z, Ebot EM, Newberg JY, Mills JM, Hegde PS, Pan Q, Dowlati A, Frampton GM, Sage J, and Lovly CM
- Subjects
- Humans, Mutation, Small Cell Lung Carcinoma pathology, Lung Neoplasms pathology, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung pathology, Carcinoma, Neuroendocrine genetics
- Abstract
Small cell lung cancer (SCLC) is a recalcitrant neuroendocrine carcinoma with dismal survival outcomes. A major barrier in the field has been the relative paucity of human tumors studied. Here we provide an integrated analysis of 3,600 "real-world" SCLC cases. This large cohort allowed us to identify new recurrent alterations and genetic subtypes, including STK11-mutant tumors (1.7%) and TP53/RB1 wild-type tumors (5.5%), as well as rare cases that were human papillomavirus-positive. In our cohort, gene amplifications on 4q12 are associated with increased overall survival, whereas CCNE1 amplification is associated with decreased overall survival. We also identify more frequent alterations in the PTEN pathway in brain metastases. Finally, profiling cases of SCLC containing oncogenic drivers typically associated with NSCLC demonstrates that SCLC transformation may occur across multiple distinct molecular cohorts of NSCLC. These novel and unsuspected genetic features of SCLC may help personalize treatment approaches for this fatal form of cancer., Significance: Minimal changes in therapy and survival outcomes have occurred in SCLC for the past four decades. The identification of new genetic subtypes and novel recurrent mutations as well as an improved understanding of the mechanisms of transformation to SCLC from NSCLC may guide the development of personalized therapies for subsets of patients with SCLC. This article is highlighted in the In This Issue feature, p. 1501., (©2023 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
18. Predicting EGFR mutational status from pathology images using a real-world dataset.
- Author
-
Pao JJ, Biggs M, Duncan D, Lin DI, Davis R, Huang RSP, Ferguson D, Janovitz T, Hiemenz MC, Eddy NR, Lehnert E, Cabili MN, Frampton GM, Hegde PS, and Albacker LA
- Subjects
- Humans, Mutation, ErbB Receptors genetics, ErbB Receptors metabolism, Retrospective Studies, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms pathology, Adenocarcinoma of Lung pathology
- Abstract
Treatment of non-small cell lung cancer is increasingly biomarker driven with multiple genomic alterations, including those in the epidermal growth factor receptor (EGFR) gene, that benefit from targeted therapies. We developed a set of algorithms to assess EGFR status and morphology using a real-world advanced lung adenocarcinoma cohort of 2099 patients with hematoxylin and eosin (H&E) images exhibiting high morphological diversity and low tumor content relative to public datasets. The best performing EGFR algorithm was attention-based and achieved an area under the curve (AUC) of 0.870, a negative predictive value (NPV) of 0.954 and a positive predictive value (PPV) of 0.410 in a validation cohort reflecting the 15% prevalence of EGFR mutations in lung adenocarcinoma. The attention model outperformed a heuristic-based model focused exclusively on tumor regions, and we show that although the attention model also extracts signal primarily from tumor morphology, it extracts additional signal from non-tumor tissue regions. Further analysis of high-attention regions by pathologists showed associations of predicted EGFR negativity with solid growth patterns and higher peritumoral immune presence. This algorithm highlights the potential of deep learning tools to provide instantaneous rule-out screening for biomarker alterations and may help prioritize the use of scarce tissue for biomarker testing., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. CD8 + T cell-intrinsic IL-6 signaling promotes resistance to anti-PD-L1 immunotherapy.
- Author
-
Huseni MA, Wang L, Klementowicz JE, Yuen K, Breart B, Orr C, Liu LF, Li Y, Gupta V, Li C, Rishipathak D, Peng J, Şenbabaoǧlu Y, Modrusan Z, Keerthivasan S, Madireddi S, Chen YJ, Fraser EJ, Leng N, Hamidi H, Koeppen H, Ziai J, Hashimoto K, Fassò M, Williams P, McDermott DF, Rosenberg JE, Powles T, Emens LA, Hegde PS, Mellman I, Turley SJ, Wilson MS, Mariathasan S, Molinero L, Merchant M, and West NR
- Subjects
- Animals, Mice, B7-H1 Antigen immunology, B7-H1 Antigen therapeutic use, CD8-Positive T-Lymphocytes metabolism, Immunotherapy, Antineoplastic Agents therapeutic use, Interleukin-6 metabolism, Neoplasms immunology, Neoplasms therapy
- Abstract
Although immune checkpoint inhibitors (ICIs) are established as effective cancer therapies, overcoming therapeutic resistance remains a critical challenge. Here we identify interleukin 6 (IL-6) as a correlate of poor response to atezolizumab (anti-PD-L1) in large clinical trials of advanced kidney, breast, and bladder cancers. In pre-clinical models, combined blockade of PD-L1 and the IL-6 receptor (IL6R) causes synergistic regression of large established tumors and substantially improves anti-tumor CD8
+ cytotoxic T lymphocyte (CTL) responses compared with anti-PD-L1 alone. Circulating CTLs from cancer patients with high plasma IL-6 display a repressed functional profile based on single-cell RNA sequencing, and IL-6-STAT3 signaling inhibits classical cytotoxic differentiation of CTLs in vitro. In tumor-bearing mice, CTL-specific IL6R deficiency is sufficient to improve anti-PD-L1 activity. Thus, based on both clinical and experimental evidence, agents targeting IL-6 signaling are plausible partners for combination with ICIs in cancer patients., Competing Interests: Declaration of interests M.A.H., K.Y., L.W., J.E.K., L.L., Y.L., V.G., C.L., D.R., C.O., S.M., S.K., Y.J.C., J.P., Y.S., Z.M., B.B., E.J.F., N.L., H.K., J.Z., M.F., P.W., M.W., I.M., S.J.T., M.M., S.M., L.M., and N.R.W. are employees of Genentech, Inc. M.A.H., K.Y., L.W., J.E.K., L.L., Y.L., P.W., M.M., S.M., L.M., and N.R.W. are inventors on patents related to IL-6. P.S.H. is an employee of Foundation Medicine Inc. K.H. is an employee of Roche Products Ltd. D.F.M. reports a consulting/advisory role for Bristol-Myers Squibb, Merck, Roche/Genentech, Pfizer, Exelixis, Novartis, Eisai, X4 Pharmaceuticals, and Array BioPharma; he also reports that his home institution receives research funding from Prometheus Laboratories. T.P. reports honoraria and consulting/advisory roles with Roche/Genentech, Bristol-Myers Squibb, and Merck; consulting/advisory role with AstraZeneca and Novartis; research funding from AstraZeneca/MedImmune and Roche/Genentech; and other relationships with Ipsen and Bristol-Myers Squibb. L.E. reports honoraria from or consulting/advisory roles with AbbVie, Amgen, AstraZeneca, Bayer, Bristol Meyers Squibb, Celgene, Chugai, eTHeRNA, Genentech, Gritstone, Medimmune, Molecuvax, Macrogenics, Novartis, Peregrine, Replimune, Roche, Silverback, Syndax, and Vaccinex; she reports that her home institution receives funding from Aduro Biotech, AstraZeneca, Breast Cancer Research Foundation, Bristol Meyers Squibb, Corvus, Department of Defense, EMD Serono, Genentech, HeritX, Inc., Maxcyte, Merck, National Cancer Institute, NSABP Foundation, Roche, Tempest, Translational Breast Cancer Research Consortium. J.E.R. has received non-financial support from Roche Genentech and consulting fees from Agensys, Eli Lilly, Sanofi, and Oncogene., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
20. Comprehensive pan-cancer genomic landscape of KRAS altered cancers and real-world outcomes in solid tumors.
- Author
-
Lee JK, Sivakumar S, Schrock AB, Madison R, Fabrizio D, Gjoerup O, Ross JS, Frampton GM, Napalkov P, Montesion M, Schutzman JL, Ye X, Hegde PS, Nagasaka M, Oxnard GR, Sokol ES, Ou SI, and Shi Z
- Abstract
Recent clinical development of KRAS inhibitors has heightened interest in the genomic landscape of KRAS-altered cancers. We performed a pan-cancer analysis of KRAS-altered samples from 426,706 adult patients with solid or hematologic malignancies using comprehensive genomic profiling; additional analyses included 62,369 liquid biopsy and 7241 pediatric samples. 23% of adult pan-cancer samples had KRAS alterations; 88% were mutations, most commonly G12D/G12V/G12C/G13D/G12R, and prevalence was similar in liquid biopsies. Co-alteration landscapes were largely similar across KRAS mutations but distinct from KRAS wild-type, though differences were observed in some tumor types for tumor mutational burden, PD-L1 expression, microsatellite instability, and other mutational signatures. Prognosis of KRAS-mutant versus other genomic cohorts of lung, pancreatic, and colorectal cancer were assessed using a real-world clinicogenomic database. As specific KRAS inhibitors and combination therapeutic strategies are being developed, genomic profiling to understand co-alterations and other biomarkers that may modulate response to targeted or immunotherapies will be imperative., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Tissue and liquid biopsy profiling reveal convergent tumor evolution and therapy evasion in breast cancer.
- Author
-
Sivakumar S, Jin DX, Tukachinsky H, Murugesan K, McGregor K, Danziger N, Pavlick D, Gjoerup O, Ross JS, Harmon R, Chung J, Decker B, Dennis L, Frampton GM, Molinero L, Oesterreich S, Venstrom JM, Oxnard GR, Hegde PS, and Sokol ES
- Subjects
- Humans, Female, Mutation, Liquid Biopsy, Biomarkers, Tumor genetics, Breast Neoplasms therapy, Breast Neoplasms drug therapy
- Abstract
Pathological and genomic profiling have transformed breast cancer care by matching patients to targeted treatments. However, tumors evolve and evade therapeutic interventions often through the acquisition of genomic mutations. Here we examine patients profiled with tissue (TBx) and liquid biopsy (LBx) as part of routine clinical care, to characterize the tumor evolutionary landscape and identify potential vulnerabilities in the relapsed setting. Real-world evidence demonstrates that LBx is utilized later in care and identifies associations with intervening therapy. While driver events are frequently shared, acquired LBx alterations are detected in a majority of patients, with the highest frequency in ER+ disease and in patients with longer biopsy intervals. Acquired mutations are often polyclonal and present at lower allelic fractions, suggesting multi-clonal convergent evolution. In addition to well-characterized resistance mutations (e.g., ESR1, NF1, RB1, ERBB2), we observe a diversity of rarer but potentially targetable mutations (e.g., PIK3CA, HRAS/NRAS/KRAS, FGFR1/2/3, BRAF) and fusions (e.g., FGFR1/2, ERBB2, RET), as well as BRCA1/2 reversions through a variety of mechanisms, including splice alterations and structural deletions. This study provides insights on treatment and selection-driven tumor evolution and identifies potential combinatorial treatment options in advanced breast cancer., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
22. Balancing speed, science and regulatory requirements in oncology drug development.
- Author
-
Snyder A, De Alwis D, Goonewardene A, and Hegde PS
- Subjects
- Drug Approval, Medical Oncology, Drug Development
- Published
- 2022
- Full Text
- View/download PDF
23. Association of CD274 (PD-L1) Copy Number Changes with Immune Checkpoint Inhibitor Clinical Benefit in Non-Squamous Non-Small Cell Lung Cancer.
- Author
-
Murugesan K, Jin DX, Comment LA, Fabrizio D, Hegde PS, Elvin JA, Alexander B, Levy MA, Frampton GM, Montesion M, Roychowdhury S, Kurzrock R, Ross JS, Albacker LA, and Huang RSP
- Subjects
- B7-H1 Antigen genetics, DNA Copy Number Variations genetics, Humans, Immune Checkpoint Inhibitors pharmacology, Immune Checkpoint Inhibitors therapeutic use, Prospective Studies, Carcinoma, Non-Small-Cell Lung drug therapy, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms drug therapy, Lung Neoplasms genetics
- Abstract
Background: We sought to characterize response to immune checkpoint inhibitor (ICI) in non-squamous non-small cell lung cancer (NSCLC) across various CD274 copy number gain and loss thresholds and identify an optimal cutoff., Materials and Methods: A de-identified nationwide (US) real-world clinico-genomic database was leveraged to study 621 non-squamous NSCLC patients treated with ICI. All patients received second-line ICI monotherapy and underwent comprehensive genomic profiling as part of routine clinical care. Overall survival (OS) from start of ICI, for CD274 copy number gain and loss cohorts across varying copy number thresholds, were assessed., Results: Among the 621 patients, patients with a CD274 CN greater than or equal to specimen ploidy +2 (N = 29) had a significantly higher median (m) OS when compared with the rest of the cohort (N = 592; 16.1 [8.9-37.3] vs 8.6 [7.1-10.9] months, hazard ratio (HR) = 0.6 [0.4-1.0], P-value = .05). Patients with a CD274 copy number less than specimen ploidy (N = 299) trended toward a lower mOS when compared to the rest of the cohort (N = 322; 7.5 [5.9-11.3] vs 9.6 [7.9-12.8] months, HR = 0.9 [0.7-1.1], P-value = .3)., Conclusion: This work shows that CD274 copy number gains at varying thresholds predict different response to ICI blockade in non-squamous NSCLC. Considering these data, prospective clinical trials should further validate these findings, specifically in the context of PD-L1 IHC test results., (© The Author(s) 2022. Published by Oxford University Press.)
- Published
- 2022
- Full Text
- View/download PDF
24. Evaluation of pre-transplant risk assessments in allogeneic hematopoietic cell transplant.
- Author
-
Hegde PS, Rybicki L, Serafino S, Bernhard L, Corrigan D, Anwer F, Kalaycio M, Sobecks RM, Jagadeesh D, Hill BT, Dean RM, Khouri J, Winter AM, Pohlman B, Majhail NS, and Hamilton BK
- Subjects
- Humans, Retrospective Studies, Risk Assessment, Transplantation, Homologous, Graft vs Host Disease, Hematopoietic Stem Cell Transplantation
- Published
- 2022
- Full Text
- View/download PDF
25. Tele-Neurorehabilitation During the COVID-19 Pandemic: Implications for Practice in Low- and Middle-Income Countries.
- Author
-
Srivastava A, Swaminathan A, Chockalingam M, Srinivasan MK, Surya N, Ray P, Hegde PS, Akkunje PS, Kamble S, Chitnis S, Kamalakannan S, Ganvir S, and Shah U
- Abstract
The importance of neurorehabilitation services for people with disabilities is getting well-recognized in low- and middle-income countries (LMICs) recently. However, accessibility to the same has remained the most significant challenge, in these contexts. This is especially because of the non-availability of trained specialists and the availability of neurorehabilitation centers only in urban cities owned predominantly by private healthcare organizations. In the current COVID-19 pandemic, the members of the Task Force for research at the Indian Federation of Neurorehabilitation (IFNR) reviewed the context for tele-neurorehabilitation (TNR) and have provided the contemporary implications for practicing TNR during COVID-19 for people with neurological disabilities (PWNDs) in LMICs. Neurorehabilitation is a science that is driven by rigorous research-based evidence. The current pandemic implies the need for systematically developed TNR interventions that is evaluated for its feasibility and acceptability and that is informed by available evidence from LMICs. Given the lack of organized systems in place for the provision of neurorehabilitation services in general, there needs to be sufficient budgetary allocations and a sector-wide approach to developing policies and systems for the provision of TNR services for PWNDs. The pandemic situation provides an opportunity to optimize the technological innovations in health and scale up these innovations to meet the growing burden of neurological disability in LMICs. Thus, this immense opportunity must be tapped to build capacity for safe and effective TNR services provision for PWNDs in these settings., 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 © 2021 Srivastava, Swaminathan, Chockalingam, Srinivasan, Surya, Ray, Hegde, Akkunje, Kamble, Chitnis, Kamalakannan, Ganvir, Shah and the Indian Federation of Neurorehabilitation (IFNR) Research Task Force.)
- Published
- 2021
- Full Text
- View/download PDF
26. Intratumoral CD103+ CD8+ T cells predict response to PD-L1 blockade.
- Author
-
Banchereau R, Chitre AS, Scherl A, Wu TD, Patil NS, de Almeida P, Kadel Iii EE, Madireddi S, Au-Yeung A, Takahashi C, Chen YJ, Modrusan Z, McBride J, Nersesian R, El-Gabry EA, Robida MD, Hung JC, Kowanetz M, Zou W, McCleland M, Caplazi P, Eshgi ST, Koeppen H, Hegde PS, Mellman I, Mathews WR, Powles T, Mariathasan S, Grogan J, and O'Gorman WE
- Subjects
- Antibodies, Monoclonal, Humanized adverse effects, B7-H1 Antigen immunology, Clinical Trials, Phase II as Topic, Clinical Trials, Phase III as Topic, Databases, Genetic, Gene Expression Profiling, Humans, Immune Checkpoint Inhibitors adverse effects, Lung Neoplasms genetics, Lung Neoplasms immunology, Phenotype, Randomized Controlled Trials as Topic, Time Factors, Treatment Outcome, Tumor Microenvironment, Urinary Bladder Neoplasms genetics, Urinary Bladder Neoplasms immunology, Antibodies, Monoclonal, Humanized therapeutic use, Antigens, CD genetics, B7-H1 Antigen antagonists & inhibitors, Biomarkers, Tumor genetics, CD8-Positive T-Lymphocytes immunology, Immune Checkpoint Inhibitors therapeutic use, Integrin alpha Chains genetics, Lung Neoplasms drug therapy, Lymphocytes, Tumor-Infiltrating immunology, Urinary Bladder Neoplasms drug therapy
- Abstract
Background: CD8+ tissue-resident memory T (T
RM ) cells, marked by CD103 ( ITGAE ) expression, are thought to actively suppress cancer progression, leading to the hypothesis that their presence in tumors may predict response to immunotherapy., Methods: Here, we test this by combining high-dimensional single-cell modalities with bulk tumor transcriptomics from 1868 patients enrolled in lung and bladder cancer clinical trials of atezolizumab (anti-programmed cell death ligand 1 (PD-L1))., Results: ITGAE was identified as the most significantly upregulated gene in inflamed tumors. Tumor CD103+ CD8+ TRM cells exhibited a complex phenotype defined by the expression of checkpoint regulators, cytotoxic proteins, and increased clonal expansion., Conclusions: Our analyses indeed demonstrate that the presence of CD103+ CD8+ TRM cells, quantified by tracking intratumoral CD103 expression, can predict treatment outcome, suggesting that patients who respond to PD-1/PD-L1 blockade are those who exhibit an ongoing antitumor T-cell response., Competing Interests: Competing interests: All authors except Thomas Powles are current or former employees of Roche., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
27. Author Correction: High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade.
- Author
-
Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, Rishipathak D, Williams P, Kadel EE 3rd, Koeppen H, Chen YJ, Modrusan Z, Grogan JL, Banchereau R, Leng N, Thastrom A, Shen X, Hashimoto K, Tayama D, van der Heijden MS, Rosenberg JE, McDermott DF, Powles T, Hegde PS, Huseni MA, and Mariathasan S
- Published
- 2021
- Full Text
- View/download PDF
28. C-reactive protein reduction post treatment is associated with improved survival in atezolizumab (anti-PD-L1) treated non-small cell lung cancer patients.
- Author
-
Patil NS, Zou W, Mocci S, Sandler A, Ballinger M, Flynn S, Kowanetz M, and Hegde PS
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Platinum therapeutic use, ROC Curve, Antibodies, Monoclonal, Humanized therapeutic use, C-Reactive Protein metabolism, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms blood, Lung Neoplasms drug therapy
- Abstract
Purpose: Overall survival (OS) is the most significant endpoint for evaluation of treatment benefit with checkpoint inhibitors (CPI) in cancer. We evaluated serum C-reactive protein (CRP) in non-small cell lung cancer (NSCLC) trials with atezolizumab (anti-PD-L1) as an early OS surrogate., Methods: Serum from patients enrolled in randomized Phase II (n = 240) and Phase III (n = 701) trials of NSCLC patients (POPLAR, OAK) who progressed on prior-platinum chemotherapy, were analyzed for CRP levels over time. Patients were grouped by changes in CRP levels post-treatment as either increased (≥ 1.5 fold), decreased (≤ 1.5 fold) or unchanged (within +1.5 fold) relative to pre-treatment levels to assess association with progression free survival (PFS) and OS., Results: Decrease in serum CRP levels at 6 weeks relative to pre-treatment were observed in patients with RECIST1.1 based complete or partial responses (CR/PR) to atezolizumab whereas patients with disease progression (PD) demonstrated an increase in CRP levels in the Phase II POPLAR study, and confirmed in the Phase III OAK study. Decrease in serum CRP as early as six weeks post treatment predicted improved PFS and OS, even in patients who were determined as stable disease (SD) in their first scan. This effect was not observed in the chemotherapy arms., Conclusion: Modulation of serum CRP correlates with clinical outcome post-atezolizumab treatment. This routine lab test may provide utility in informing OS signals as early as 6 weeks post-initiation of therapy with CPIs in NSCLC., Competing Interests: The authors have read the journal’s policy and the authors of this paper declare the following competing interests: Genentech Roche provided support in the form of salaries for authors [NSP, WZ, SM, AS, MB, SF, MK, PSH]. There are no patents, products in development or marketed products associated with this research to declare. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
29. A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases.
- Author
-
Huang RSP, Haberberger J, Severson E, Duncan DL, Hemmerich A, Edgerly C, Ferguson NL, Williams E, Elvin J, Vergilio JA, Killian JK, Lin DI, Tse J, Hiemenz M, Owens C, Danziger N, Hegde PS, Venstrom J, Alexander B, Ross JS, and Ramkissoon SH
- Subjects
- B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Gene Amplification, Humans, Immunohistochemistry, Microsatellite Instability, Mutation, Retrospective Studies, B7-H1 Antigen analysis, Biomarkers, Tumor analysis, Neoplasms genetics, Neoplasms metabolism
- Abstract
PD-L1 immunohistochemistry (IHC) currently has the most Food and Drug Administration (FDA) approvals as a companion diagnostic (CDx) for immunotherapies in specific tumor types; however, multiple other immunotherapy biomarkers exist. We performed this study to examine and report the prevalence of PD-L1 expression in a wide variety of tumor types and examine its relationship to microsatellite instability (MSI), tumor mutational burden (TMB), and CD274 (PD-L1) gene amplification. We performed a retrospective analysis of all cases in which both PD-L1 IHC (using the DAKO 22C3 IHC assay with either tumor proportion score (TPS) or combined positive score (CPS); or the VENTANA SP142 assay with infiltrating immune cell score (IC)) and comprehensive genomic profiling (CGP) were tested at Foundation Medicine between January 2016 and November 2019. Of note, PD-L1 positivity is defined per the CDx indication and tumor proportion score (TPS ≥ 1) for indications without a CDx claim; and TMB positivity is defined as ≥10 mutations/Mb. A total of 48,782 cases were tested for PD-L1 IHC and CGP. Immune cell expression of PD-L1 was more frequently identified than tumor cell expression of PD-L1. We saw a high correlation between PD-L1 expression and CD274 gene amplification (p < 0.0001), MSI and TMB (p < 0.0001), and PD-L1 and TMB (p < 0.0001). In addition, the combination of PD-L1 and TMB identified four unique disease subsets PD-L1
- /TMB- , PD-L1+ /TMB- , PD-L1- /TMB+ , and PD-L1+ /TMB+ with varying prevalence dependent on tumor type. Lastly, 50.3% (24527/48782) of the overall cohort was positive for at least one of the CDx or exploratory biomarkers described above. This is the largest pan-cancer analysis of relevant biomarkers associated with response to checkpoint inhibitors to date, including more than 48,000 cases. Additional clinical trials with treatment outcome data in individual tumor types are needed to determine whether the double positive PD-L1+ /TMB+ disease subset would respond best to immunotherapy.- Published
- 2021
- Full Text
- View/download PDF
30. Somatic HLA Class I Loss Is a Widespread Mechanism of Immune Evasion Which Refines the Use of Tumor Mutational Burden as a Biomarker of Checkpoint Inhibitor Response.
- Author
-
Montesion M, Murugesan K, Jin DX, Sharaf R, Sanchez N, Guria A, Minker M, Li G, Fisher V, Sokol ES, Pavlick DC, Moore JA, Braly A, Singal G, Fabrizio D, Comment LA, Rizvi NA, Alexander BM, Frampton GM, Hegde PS, and Albacker LA
- Subjects
- Biomarkers, Tumor genetics, Humans, Mutation, Tumor Escape, Carcinoma, Non-Small-Cell Lung drug therapy, HLA Antigens genetics, Immune Checkpoint Inhibitors therapeutic use, Lung Neoplasms drug therapy
- Abstract
Neoantigen presentation arises as a result of tumor-specific mutations and is a critical component of immune surveillance that can be abrogated by somatic LOH of the human leukocyte antigen class I (HLA-I) locus. To understand the role of HLA-I LOH in oncogenesis and treatment, we utilized a pan-cancer genomic dataset of 83,644 patient samples, a small subset of which had treatment outcomes with immune checkpoint inhibitors (ICI). HLA-I LOH was common (17%) and unexpectedly had a nonlinear relationship with tumor mutational burden (TMB). HLA-I LOH was frequent at intermediate TMB, yet prevalence decreased above 30 mutations/megabase, suggesting highly mutated tumors require alternate immune evasion mechanisms. In ICI-treated patients with nonsquamous non-small cell lung cancer, HLA-I LOH was a significant negative predictor of overall survival. Survival prediction improved when combined with TMB, suggesting TMB with HLA-I LOH may better identify patients likely to benefit from ICIs. SIGNIFICANCE: This work shows the pan-cancer landscape of HLA-I LOH, revealing an unexpected "Goldilocks" relationship between HLA-I LOH and TMB, and demonstrates HLA-I LOH as a significant negative predictor of outcomes after ICI treatment. These data informed a combined predictor of outcomes after ICI and have implications for tumor vaccine development. This article is highlighted in the In This Issue feature, p. 211 ., (©2020 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
31. Molecular Subsets in Renal Cancer Determine Outcome to Checkpoint and Angiogenesis Blockade.
- Author
-
Motzer RJ, Banchereau R, Hamidi H, Powles T, McDermott D, Atkins MB, Escudier B, Liu LF, Leng N, Abbas AR, Fan J, Koeppen H, Lin J, Carroll S, Hashimoto K, Mariathasan S, Green M, Tayama D, Hegde PS, Schiff C, Huseni MA, and Rini B
- Subjects
- Angiogenesis Inhibitors pharmacology, Antibodies, Monoclonal, Humanized pharmacology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bevacizumab pharmacology, Bevacizumab therapeutic use, Carcinoma, Renal Cell genetics, Clinical Trials, Phase III as Topic, Computational Biology methods, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Humans, Immune Checkpoint Inhibitors pharmacology, Kidney Neoplasms genetics, Prognosis, Randomized Controlled Trials as Topic, Sequence Analysis, RNA, Sunitinib pharmacology, Sunitinib therapeutic use, Treatment Outcome, Unsupervised Machine Learning, Angiogenesis Inhibitors therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor genetics, Carcinoma, Renal Cell drug therapy, Immune Checkpoint Inhibitors therapeutic use, Kidney Neoplasms drug therapy
- Abstract
Integrated multi-omics evaluation of 823 tumors from advanced renal cell carcinoma (RCC) patients identifies molecular subsets associated with differential clinical outcomes to angiogenesis blockade alone or with a checkpoint inhibitor. Unsupervised transcriptomic analysis reveals seven molecular subsets with distinct angiogenesis, immune, cell-cycle, metabolism, and stromal programs. While sunitinib and atezolizumab + bevacizumab are effective in subsets with high angiogenesis, atezolizumab + bevacizumab improves clinical benefit in tumors with high T-effector and/or cell-cycle transcription. Somatic mutations in PBRM1 and KDM5C associate with high angiogenesis and AMPK/fatty acid oxidation gene expression, while CDKN2A/B and TP53 alterations associate with increased cell-cycle and anabolic metabolism. Sarcomatoid tumors exhibit lower prevalence of PBRM1 mutations and angiogenesis markers, frequent CDKN2A/B alterations, and increased PD-L1 expression. These findings can be applied to molecularly stratify patients, explain improved outcomes of sarcomatoid tumors to checkpoint blockade versus antiangiogenics alone, and develop personalized therapies in RCC and other indications., Competing Interests: Declaration of Interests R.J.M. has received honoraria for advisory roles from Roche–Genentech (RGE), Pfizer, Novartis, Exelixis, Eisai, Lilly Oncology, AstraZeneca (AZ), Incyte, and Merck Sharp & Dohme (MSD), and institutional support from Bristol-Myers Squibb (BMS), RGE, Pfizer, Novartis, Exelixis, and Eisai outside of the submitted work. T.P. has received grants from AZ and RGE, and honoraria from AZ, RGE, BMS, Pfizer, Novartis, Exelixis, and MSD outside of the submitted work. DFM has received grants from BMS and Prometheus and honoraria for consulting roles from BMS, Pfizer, MSD, Novartis, Eisai, Exelixis, Array BioPharm, and RGE outside of the submitted work. M.B.A. has received grants from RGE during the conduct of the study and honoraria for consulting roles outside of the submitted work from RGE, BMS, MSD, Novartis, Pfizer, Exelixis, and Esai. B.E. has received grants and honoraria from BMS, Novartis, Ipsen, and EUSA outside of the submitted work. B.R. has received grants and honoraria from RGE and Pfizer during the conduct of the study and has received grants to his institution from MSD, Peloton, Aveo, BMS, AZ, and honoraria for consulting roles from Novartis, Synthorx, Compugen, Corvus, Exelixis, Arravive, Surface Oncology, 3D Medicines, and holds stock in PTC therapeutics, all outside of the submitted work. R.B., H.H., L.-F.L., N.L., A.R.A., J.F., H.K., J.L., S.M., M.G., D.T., and M.A.H. are employees of Genentech Inc. S.C. is an employee of Calithera Biosciences Inc. K.H. is an employee of Crescendo Biologics Inc. P.S.H. is an employee of Foundation Medicine Inc., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
32. Evaluating the Psychometric Properties of the Kannada Version of EAT 10.
- Author
-
Krishnamurthy R, Balasubramanium RK, and Hegde PS
- Subjects
- Humans, Psychometrics, Reproducibility of Results, Surveys and Questionnaires, Deglutition Disorders diagnosis, Language
- Abstract
Background: Eating Assessment Tool 10 is a symptom survey and has several advantages over other existing questionnaires. The instrument has also proven to be useful in establishing initial dysphagia symptom severity and to aid in measuring treatment outcomes. Due to its wide applicability, the instrument has been translated into several languages. The aim of the present study was to translate and validate the Kannada version of Eating Assessment Tool 10., Method: 88 individuals with dysphagia and equal number of healthy individuals filled the Kannada version of Eating Assessment Tool. Internal consistency and test-retest reproducibility were used for reliability testing. Validity was also established by comparing the scores of dysphagia patients and healthy controls., Results: The overall Cronbach's α for the tool was 0.9 indicating a good internal consistency. The internal consistency of each of the items was also high, and ranged 0.88 to 0.9. Twenty patients filled the questionnaire after a span of 48 h, and the ICC coefficient was found to be 0.89 indicating a high reliability. The control group has significantly lower scores for all scales when compared to the dysphagia group [t(174) = 78.41, p < 0.001]., Conclusion: The present study demonstrates that the Kannada version of EAT 10 has good internal consistency, test retest reliability, and concurrent validity. The results of the study also reveal that it is a reliable and valid tool for screening dysphagia population.
- Published
- 2020
- Full Text
- View/download PDF
33. Gustatory dysfunction in non oral irradiation for head and neck cancers.
- Author
-
Hegde PS, Aanya A, Joshna BM, Thakur S, and Rao V
- Subjects
- Autonomic Nervous System Diseases diagnosis, Female, Head and Neck Neoplasms diagnosis, Head and Neck Neoplasms radiotherapy, Humans, Male, Organs at Risk, Radiotherapy methods, Autonomic Nervous System Diseases etiology, Autonomic Nervous System Diseases physiopathology, Head and Neck Neoplasms complications, Radiotherapy adverse effects, Taste
- Abstract
Competing Interests: Declaration of Competing Interest The authors declared that there is no conflict of interest.
- Published
- 2020
- Full Text
- View/download PDF
34. Evidence-Based Development and Clinical Use of Precision Oncology Therapeutics.
- Author
-
Comment LA, Ward AF, Schrock AB, Fabrizio D, Venstrom JM, Hegde PS, and Alexander BM
- Subjects
- Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Clinical Decision-Making, Humans, Molecular Diagnostic Techniques, Molecular Targeted Therapy, Neoplasms genetics, Neoplasms metabolism, Antineoplastic Agents therapeutic use, Drug Development, Evidence-Based Medicine, Neoplasms drug therapy, Precision Medicine
- Published
- 2020
- Full Text
- View/download PDF
35. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade.
- Author
-
Yuen KC, Liu LF, Gupta V, Madireddi S, Keerthivasan S, Li C, Rishipathak D, Williams P, Kadel EE 3rd, Koeppen H, Chen YJ, Modrusan Z, Grogan JL, Banchereau R, Leng N, Thastrom A, Shen X, Hashimoto K, Tayama D, van der Heijden MS, Rosenberg JE, McDermott DF, Powles T, Hegde PS, Huseni MA, and Mariathasan S
- Subjects
- Adult, Antibodies, Monoclonal, Humanized therapeutic use, B7-H1 Antigen immunology, Biomarkers, Pharmacological blood, Biomarkers, Pharmacological metabolism, Biomarkers, Tumor blood, Biomarkers, Tumor metabolism, Carcinoma, Renal Cell diagnosis, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell mortality, Carcinoma, Transitional Cell diagnosis, Carcinoma, Transitional Cell drug therapy, Carcinoma, Transitional Cell metabolism, Carcinoma, Transitional Cell mortality, Drug Resistance, Neoplasm, Female, Humans, Interleukin-8 blood, Kidney Neoplasms diagnosis, Kidney Neoplasms drug therapy, Kidney Neoplasms metabolism, Kidney Neoplasms mortality, Male, Neoplasms metabolism, Neoplasms mortality, Prognosis, Survival Analysis, Treatment Failure, Urologic Neoplasms diagnosis, Urologic Neoplasms drug therapy, Urologic Neoplasms metabolism, Urologic Neoplasms mortality, Antineoplastic Agents, Immunological therapeutic use, B7-H1 Antigen antagonists & inhibitors, Interleukin-8 metabolism, Neoplasms diagnosis, Neoplasms drug therapy
- Abstract
Although elevated plasma interleukin-8 (pIL-8) has been associated with poor outcome to immune checkpoint blockade
1 , this has not been comprehensively evaluated in large randomized studies. Here we analyzed circulating pIL-8 and IL8 gene expression in peripheral blood mononuclear cells and tumors of patients treated with atezolizumab (anti-PD-L1 monoclonal antibody) from multiple randomized trials representing 1,445 patients with metastatic urothelial carcinoma (mUC) and metastatic renal cell carcinoma. High levels of IL-8 in plasma, peripheral blood mononuclear cells and tumors were associated with decreased efficacy of atezolizumab in patients with mUC and metastatic renal cell carcinoma, even in tumors that were classically CD8+ T cell inflamed. Low baseline pIL-8 in patients with mUC was associated with increased response to atezolizumab and chemotherapy. Patients with mUC who experienced on-treatment decreases in pIL-8 exhibited improved overall survival when treated with atezolizumab but not with chemotherapy. Single-cell RNA sequencing of the immune compartment showed that IL8 is primarily expressed in circulating and intratumoral myeloid cells and that high IL8 expression is associated with downregulation of the antigen-presentation machinery. Therapies that can reverse the impacts of IL-8-mediated myeloid inflammation will be essential for improving outcomes of patients treated with immune checkpoint inhibitors.- Published
- 2020
- Full Text
- View/download PDF
36. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy.
- Author
-
Mayoux M, Roller A, Pulko V, Sammicheli S, Chen S, Sum E, Jost C, Fransen MF, Buser RB, Kowanetz M, Rommel K, Matos I, Colombetti S, Belousov A, Karanikas V, Ossendorp F, Hegde PS, Chen DS, Umana P, Perro M, Klein C, and Xu W
- Subjects
- Dendritic Cells, Humans, Antibodies, Monoclonal, Humanized therapeutic use, B7-H1 Antigen antagonists & inhibitors, Carcinoma, Non-Small-Cell Lung drug therapy, Immunotherapy, Lung Neoplasms drug therapy
- Abstract
PD-L1/PD-1 blocking antibodies have demonstrated therapeutic efficacy across a range of human cancers. Extending this benefit to a greater number of patients, however, will require a better understanding of how these therapies instigate anticancer immunity. Although the PD-L1/PD-1 axis is typically associated with T cell function, we demonstrate here that dendritic cells (DCs) are an important target of PD-L1 blocking antibody. PD-L1 binds two receptors, PD-1 and B7.1 (CD80). PD-L1 is expressed much more abundantly than B7.1 on peripheral and tumor-associated DCs in patients with cancer. Blocking PD-L1 on DCs relieves B7.1 sequestration in cis by PD-L1, which allows the B7.1/CD28 interaction to enhance T cell priming. In line with this, in patients with renal cell carcinoma or non-small cell lung cancer treated with atezolizumab (PD-L1 blockade), a DC gene signature is strongly associated with improved overall survival. These data suggest that PD-L1 blockade reinvigorates DC function to generate potent anticancer T cell immunity., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
37. Intolerance to pungent spices among patients receiving radiation and chemotherapy for head and neck cancers.
- Author
-
Aanya A, Hegde PS, Fernando CA, Thakur S, and Rao V
- Subjects
- Food Intolerance etiology, Head and Neck Neoplasms pathology, Humans, Prognosis, Chemoradiotherapy adverse effects, Food Intolerance pathology, Head and Neck Neoplasms therapy, Irritants adverse effects, Spices adverse effects
- Abstract
Competing Interests: Declaration of competing interest: The authors state that they have no known competing for financial interests or personal relationship of persons or organizations that could have appeared to influence the writing in this paper.
- Published
- 2020
- Full Text
- View/download PDF
38. Top 10 Challenges in Cancer Immunotherapy.
- Author
-
Hegde PS and Chen DS
- Subjects
- Humans, Models, Biological, Neoplasms genetics, Neoplasms immunology, Tumor Microenvironment immunology, Immunotherapy methods, Neoplasms therapy
- Abstract
Cancer immunotherapy is a validated and critically important approach for treating patients with cancer. Given the vast research and clinical investigation efforts dedicated to advancing both endogenous and synthetic immunotherapy approaches, there is a need to focus on crucial questions and define roadblocks to the basic understanding and clinical progress. Here, we define ten key challenges facing cancer immunotherapy, which range from lack of confidence in translating pre-clinical findings to identifying optimal combinations of immune-based therapies for any given patient. Addressing these challenges will require the combined efforts of basic researchers and clinicians, and the focusing of resources to accelerate understanding of the complex interactions between cancer and the immune system and the development of improved treatment options for patients with cancer., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
39. A Novel Next-Generation Sequencing Approach to Detecting Microsatellite Instability and Pan-Tumor Characterization of 1000 Microsatellite Instability-High Cases in 67,000 Patient Samples.
- Author
-
Trabucco SE, Gowen K, Maund SL, Sanford E, Fabrizio DA, Hall MJ, Yakirevich E, Gregg JP, Stephens PJ, Frampton GM, Hegde PS, Miller VA, Ross JS, Hartmaier RJ, Huang SA, and Sun JX
- Subjects
- Algorithms, Female, Gene Frequency, High-Throughput Nucleotide Sequencing statistics & numerical data, Humans, Male, Mutation, Principal Component Analysis, High-Throughput Nucleotide Sequencing methods, Microsatellite Instability, Neoplasms genetics
- Abstract
Microsatellite instability (MSI) is an important biomarker for predicting response to immune checkpoint inhibitor therapy, as emphasized by the recent checkpoint inhibitor approval for MSI-high (MSI-H) solid tumors. Herein, we describe and validate a novel method for determining MSI status from a next-generation sequencing comprehensive genomic profiling assay using formalin-fixed, paraffin-embedded samples. This method is 97% (65/67) concordant with current standards, PCR and immunohistochemistry. We further apply this method to >67,000 patient tumor samples to identify genes and pathways that are enriched in MSI-stable or MSI-H tumor groups. Data show that although rare in tumors other than colorectal and endometrial carcinomas, MSI-H samples are present in many tumor types. Furthermore, the large sample set revealed that MSI-H tumors selectively share alterations in genes across multiple common pathways, including WNT, phosphatidylinositol 3-kinase, and NOTCH. Last, MSI is sufficient, but not necessary, for a tumor to have elevated tumor mutation burden. Therefore, MSI can be determined from comprehensive genomic profiling with high accuracy, allowing for efficient MSI-H detection across all tumor types, especially those in which routine use of immunohistochemistry or PCR-based assays would be impractical because of a rare incidence of MSI. MSI-H tumors are enriched in alterations in specific signaling pathways, providing a rationale for investigating directed immune checkpoint inhibitor therapies in combination with pathway-targeted therapies., (Copyright © 2019 American Society for Investigative Pathology and the Association for Molecular Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
40. Safety, Clinical Activity, and Biological Correlates of Response in Patients with Metastatic Melanoma: Results from a Phase I Trial of Atezolizumab.
- Author
-
Hamid O, Molinero L, Bolen CR, Sosman JA, Muñoz-Couselo E, Kluger HM, McDermott DF, Powderly JD, Sarkar I, Ballinger M, Fassò M, O'Hear C, Chen DS, Hegde PS, and Hodi FS
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized administration & dosage, Antineoplastic Agents, Immunological administration & dosage, B7-H1 Antigen analysis, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen immunology, Biomarkers, Tumor immunology, Fatigue chemically induced, Fatigue diagnosis, Female, Fever chemically induced, Fever diagnosis, Follow-Up Studies, Humans, Male, Melanoma immunology, Melanoma mortality, Melanoma secondary, Middle Aged, Prognosis, Progression-Free Survival, Pruritus chemically induced, Pruritus diagnosis, Retrospective Studies, Severity of Illness Index, Skin immunology, Skin pathology, Skin Neoplasms immunology, Skin Neoplasms mortality, Skin Neoplasms pathology, T-Lymphocytes, Cytotoxic immunology, Transcriptome, Young Adult, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological adverse effects, Biomarkers, Tumor analysis, Melanoma drug therapy, Skin Neoplasms drug therapy
- Abstract
Purpose: Atezolizumab [anti-programmed death-ligand 1 (PD-L1)] selectively targets PD-L1 to block its interaction with receptors programmed death 1 and B7.1, thereby reinvigorating antitumor T-cell activity. We evaluated the long-term safety and activity of atezolizumab, along with biological correlates of clinical activity endpoints, in a cohort of patients with melanoma in an ongoing phase Ia study (NCT01375842)., Patients and Methods: Patients with unresectable or metastatic melanoma were enrolled to receive atezolizumab 0.1 to 20 mg/kg or ≥10 mg/kg every 3 weeks. Primary study objectives were safety and tolerability. Secondary objectives included investigator-assessed efficacy measures; pharmacodynamic and predictive biomarkers of antitumor activity were explored., Results: Forty-five patients were enrolled and were evaluable for safety. Most treatment-related adverse events (AE) were grade 1/2 (60%). Fatigue (44%), pruritus (20%), pyrexia (18%), and rash (18%) were the most common treatment-related AEs of any grade. No treatment-related deaths occurred. Overall response rate was 30% among 43 efficacy- evaluable patients, with a median duration of response of 62 months [95% CI, 35-not estimable (NE)]. Clinically meaningful long-term survival was observed, with a median overall survival of 23 months (95% CI, 9-66). Baseline biomarkers of tumor immunity [PD-L1 expression on immune cells, T effector (Teff), and antigen presentation gene signatures) and tumor mutational burden (TMB) were associated with improved response, progression-free survival, and overall survival., Conclusions: Atezolizumab was well tolerated, with durable responses and survival in patients with melanoma. PD-L1 expression, TMB, and Teff signatures may indicate improved benefit with atezolizumab in these patients., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
41. Late-differentiated effector neoantigen-specific CD8+ T cells are enriched in peripheral blood of non-small cell lung carcinoma patients responding to atezolizumab treatment.
- Author
-
Fehlings M, Jhunjhunwala S, Kowanetz M, O'Gorman WE, Hegde PS, Sumatoh H, Lee BH, Nardin A, Becht E, Flynn S, Ballinger M, Newell EW, and Yadav M
- Subjects
- Aged, Antibodies, Monoclonal, Humanized pharmacology, Antigens, Neoplasm genetics, Antigens, Neoplasm isolation & purification, Antigens, Neoplasm metabolism, Antineoplastic Agents, Immunological pharmacology, B7-H1 Antigen antagonists & inhibitors, B7-H1 Antigen immunology, CD8-Positive T-Lymphocytes metabolism, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung immunology, Drug Monitoring methods, Female, Humans, Lung Neoplasms blood, Lung Neoplasms genetics, Lung Neoplasms immunology, Male, Middle Aged, Mutation, RNA-Seq, Exome Sequencing, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, CD8-Positive T-Lymphocytes immunology, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy
- Abstract
Background: There is strong evidence that immunotherapy-mediated tumor rejection can be driven by tumor-specific CD8+ T cells reinvigorated to recognize neoantigens derived from tumor somatic mutations. Thus, the frequencies or characteristics of tumor-reactive, mutation-specific CD8+ T cells could be used as biomarkers of an anti-tumor response. However, such neoantigen-specific T cells are difficult to reliably identify due to their low frequency in peripheral blood and wide range of potential epitope specificities., Methods: Peripheral blood mononuclear cells (PBMC) from 14 non-small cell lung cancer (NSCLC) patients were collected pre- and post-treatment with the anti-PD-L1 antibody atezolizumab. Using whole exome sequencing and RNA sequencing we identified tumor neoantigens that are predicted to bind to major histocompatibility complex class I (MHC-I) and utilized mass cytometry, together with cellular 'barcoding', to profile immune cells from patients with objective response to therapy (n = 8) and those with progressive disease (n = 6). In parallel, a highly-multiplexed combinatorial tetramer staining was used to screen antigen-specific CD8+ T cells in peripheral blood for 782 candidate tumor neoantigens and 71 known viral-derived control peptide epitopes across all patient samples., Results: No significant treatment- or response associated phenotypic difference were measured in bulk CD8+ T cells. Multiplexed peptide-MHC multimer staining detected 20 different neoantigen-specific T cell populations, as well as T cells specific for viral control antigens. Not only were neoantigen-specific T cells more frequently detected in responding patients, their phenotypes were also almost entirely distinct. Neoantigen-specific T cells from responder patients typically showed a differentiated effector phenotype, most like Cytomegalovirus (CMV) and some types of Epstein-Barr virus (EBV)-specific CD8+ T cells. In contrast, more memory-like phenotypic profiles were observed for neoantigen-specific CD8+ T cells from patients with progressive disease., Conclusion: This study demonstrates that neoantigen-specific T cells can be detected in peripheral blood in non-small cell lung cancer (NSCLC) patients during anti-PD-L1 therapy. Patients with an objective response had an enrichment of neoantigen-reactive T cells and these cells showed a phenotype that differed from patients without a response. These findings suggest the ex vivo identification, characterization, and longitudinal follow-up of rare tumor-specific differentiated effector neoantigen-specific T cells may be useful in predicting response to checkpoint blockade., Trial Registration: POPLAR trial NCT01903993 .
- Published
- 2019
- Full Text
- View/download PDF
42. Development of a PD-L1 Complementary Diagnostic Immunohistochemistry Assay (SP142) for Atezolizumab.
- Author
-
Vennapusa B, Baker B, Kowanetz M, Boone J, Menzl I, Bruey JM, Fine G, Mariathasan S, McCaffery I, Mocci S, Rost S, Smith D, Dennis E, Tang SY, Damadzadeh B, Walker E, Hegde PS, Williams JA, Koeppen H, and Boyd Z
- Subjects
- B7-H1 Antigen antagonists & inhibitors, Carcinoma, Non-Small-Cell Lung immunology, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms immunology, Observer Variation, Patient Selection, Reproducibility of Results, Sensitivity and Specificity, Urinary Bladder Neoplasms immunology, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, B7-H1 Antigen metabolism, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung therapy, Immunohistochemistry methods, Immunotherapy methods, Lung Neoplasms therapy, Urinary Bladder Neoplasms therapy
- Abstract
Cancer immunotherapies, such as atezolizumab, are proving to be a valuable therapeutic strategy across indications, including non-small cell lung cancer (NSCLC) and urothelial cancer (UC). Here, we describe a diagnostic assay that measures programmed-death ligand 1 (PD-L1) expression, via immunohistochemistry, to identify patients who will derive the most benefit from treatment with atezolizumab, a humanized monoclonal anti-PD-L1 antibody. We describe the performance of the VENTANA PD-L1 (SP142) Assay in terms of specificity, sensitivity, and the ability to stain both tumor cells (TC) and tumor-infiltrating immune cells (IC), in NSCLC and UC tissues. The reader precision, repeatability and intermediate precision, interlaboratory reproducibility, and the effectiveness of pathologist training on the assessment of PD-L1 staining on both TC and IC were evaluated. We detail the analytical validation of the VENTANA PD-L1 (SP142) Assay for PD-L1 expression in NSCLC and UC tissues and show that the assay reliably evaluated staining on both TC and IC across multiple expression levels/clinical cut-offs. The reader precision showed high overall agreement when compared with consensus scores. In addition, pathologists met the predefined training criteria (≥85.0% overall percent agreement) for the assessment of PD-L1 expression in NSCLC and UC tissues with an average overall percent agreement ≥95.0%. The assay evaluates PD-L1 staining on both cell types and is robust and precise. In addition, it can help to identify those patients who may benefit the most from treatment with atezolizumab, although treatment benefit has been demonstrated in an all-comer NSCLC and UC patient population.
- Published
- 2019
- Full Text
- View/download PDF
43. Publisher Correction: Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma.
- Author
-
McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, Fong L, Joseph RW, Pal SK, Reeves JA, Sznol M, Hainsworth J, Rathmell WK, Stadler WM, Hutson T, Gore ME, Ravaud A, Bracarda S, Suárez C, Danielli R, Gruenwald V, Choueiri TK, Nickles D, Jhunjhunwala S, Piault-Louis E, Thobhani A, Qiu J, Chen DS, Hegde PS, Schiff C, Fine GD, and Powles T
- Abstract
In the version of this article originally published, there was an error in Fig. 2n. The top line of the HR comparison chart originally was Atezo + bev vs sun. It should have been Atezo + bev vs atezo. The error has been corrected in the HTML and PDF versions of this article.
- Published
- 2018
- Full Text
- View/download PDF
44. Transcriptomic analysis of hepatocellular carcinoma reveals molecular features of disease progression and tumor immune biology.
- Author
-
Okrah K, Tarighat S, Liu B, Koeppen H, Wagle MC, Cheng G, Sun C, Dey A, Chang MT, Sumiyoshi T, Mounir Z, Cummings C, Hampton G, Amler L, Fridlyand J, Hegde PS, Turley SJ, Lackner MR, and Huang SM
- Abstract
Hepatocellular carcinoma (HCC) develops in the context of chronic inflammatory liver disease and has an extremely poor prognosis. An immunosuppressive tumor microenvironment may contribute to therapeutic failure in metastatic HCC. Here, we identified unique molecular signatures pertaining to HCC disease progression and tumor immunity by analyzing genome-wide RNA-Seq data derived from HCC patient tumors and non-tumor cirrhotic tissues. Unsupervised clustering of gene expression data revealed a gradual suppression of local tumor immunity that coincided with disease progression, indicating an increasingly immunosuppressive tumor environment during HCC disease advancement. IHC examination of the spatial distribution of CD8+ T cells in tumors revealed distinct intra- and peri-tumoral subsets. Differential gene expression analysis revealed an 85-gene signature that was significantly upregulated in the peri-tumoral CD8+ T cell-excluded tumors. Notably, this signature was highly enriched with components of underlying extracellular matrix, fibrosis, and epithelial-mesenchymal transition (EMT). Further analysis condensed this signature to a core set of 23 genes that are associated with CD8+ T cell localization, and were prospectively validated in an independent cohort of HCC specimens. These findings suggest a potential association between elevated fibrosis, possibly modulated by TGF-β, PDGFR, SHH or Notch pathway, and the T cell-excluded immune phenotype. Indeed, targeting fibrosis using a TGF-β neutralizing antibody in the STAM™ model of murine HCC, we found that ameliorating the fibrotic environment could facilitate redistribution of CD8+ lymphocytes into tumors. Our results provide a strong rationale for utilizing immunotherapies in HCC earlier during treatment, potentially in combination with anti-fibrotic therapies., Competing Interests: All authors are current or former employees and shareholders of Roche/Genentech.
- Published
- 2018
- Full Text
- View/download PDF
45. Differential regulation of PD-L1 expression by immune and tumor cells in NSCLC and the response to treatment with atezolizumab (anti-PD-L1).
- Author
-
Kowanetz M, Zou W, Gettinger SN, Koeppen H, Kockx M, Schmid P, Kadel EE 3rd, Wistuba I, Chaft J, Rizvi NA, Spigel DR, Spira A, Hirsch FR, Cohen V, Smith D, Boyd Z, Miley N, Flynn S, Leveque V, Shames DS, Ballinger M, Mocci S, Shankar G, Funke R, Hampton G, Sandler A, Amler L, Mellman I, Chen DS, and Hegde PS
- Subjects
- Antibodies, Monoclonal, Humanized, Humans, Immunohistochemistry methods, Lung Neoplasms drug therapy, Lymphocytes, Tumor-Infiltrating drug effects, Male, Middle Aged, T-Lymphocytes drug effects, T-Lymphocytes immunology, Antibodies, Monoclonal therapeutic use, B7-H1 Antigen immunology, Carcinoma, Non-Small-Cell Lung immunology, Lung Neoplasms immunology
- Abstract
Programmed death-ligand 1 (PD-L1) expression on tumor cells (TCs) by immunohistochemistry is rapidly gaining importance as a diagnostic for the selection or stratification of patients with non-small cell lung cancer (NSCLC) most likely to respond to single-agent checkpoint inhibitors. However, at least two distinct patterns of PD-L1 expression have been observed with potential biological and clinical relevance in NSCLC: expression on TC or on tumor-infiltrating immune cells (ICs). We investigated the molecular and cellular characteristics associated with PD-L1 expression in these distinct cell compartments in 4,549 cases of NSCLC. PD-L1 expression on IC was more prevalent and likely reflected IFN-γ-induced adaptive regulation accompanied by increased tumor-infiltrating lymphocytes and effector T cells. High PD-L1 expression on TC, however, reflected an epigenetic dysregulation of the PD-L1 gene and was associated with a distinct histology described by poor immune infiltration, sclerotic/desmoplastic stroma, and mesenchymal molecular features. Importantly, durable clinical responses to atezolizumab (anti-PD-L1) were observed in patients with tumors expressing high PD-L1 levels on either TC alone [40% objective response rate (ORR)] or IC alone (22% ORR). Thus, PD-L1 expression on TC or IC can independently attenuate anticancer immunity and emphasizes the functional importance of IC in regulating the antitumor T cell response., Competing Interests: Conflict of interest statement: M. Kowanetz, W.Z., H.K., E.E.K., D.S., Z.B., N.M., S.F., V.L., D.S.S., M.B., S.M., G.S., R.F., G.H., A. Sandler, L.A., I.M., D.S.C., and P.S.H. are full-time employees of Genentech, which supported the research described and markets the therapeutic antibody atezolizumab, under study., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
46. Predictive markers of anti-VEGF and emerging role of angiogenesis inhibitors as immunotherapeutics.
- Author
-
Hegde PS, Wallin JJ, and Mancao C
- Subjects
- Angiogenesis Inhibitors immunology, Humans, Immunotherapy methods, Angiogenesis Inhibitors therapeutic use, Biomarkers, Tumor immunology, Neoplasms drug therapy, Neoplasms immunology, Neovascularization, Pathologic drug therapy, Neovascularization, Pathologic immunology, Vascular Endothelial Growth Factor A immunology
- Abstract
The critical role of angiogenesis in promoting tumor growth and metastasis has been well established scientifically, and consequently blocking this pathway as a therapeutic strategy has demonstrated great clinical success for the treatment of cancer. The holy grail however, has been the identification of patients who derive significant survival benefit from this class of agents. Here we attempt to delineate the diverse mechanisms related to anti-VEGF including its role as an anti-vascular, anti-angiogenic or an anti-permeability factor and review the most promising predictive biomarkers interrogated in large clinical trials, that identify patients who may derive significant survival advantage with VEGF inhibition. Lastly, we describe the function of VEGF as an immunomodulator and illustrate the evidence for anti-VEGF in reprogramming the tumor milieu from an immunosuppressive to an immune permissive microenvironment in human cancers, thus elucidating the role of anti-VEGF as an optimal combination partner for immune checkpoint inhibitors., (Copyright © 2017 Elsevier Ltd. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
47. Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab.
- Author
-
Gandara DR, Paul SM, Kowanetz M, Schleifman E, Zou W, Li Y, Rittmeyer A, Fehrenbacher L, Otto G, Malboeuf C, Lieber DS, Lipson D, Silterra J, Amler L, Riehl T, Cummings CA, Hegde PS, Sandler A, Ballinger M, Fabrizio D, Mok T, and Shames DS
- Subjects
- Antibodies, Monoclonal, Humanized, B7-H1 Antigen metabolism, Biomarkers, Tumor metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Humans, Immunotherapy, Kaplan-Meier Estimate, Lung Neoplasms drug therapy, Progression-Free Survival, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Carcinoma, Non-Small-Cell Lung blood, Carcinoma, Non-Small-Cell Lung genetics, Lung Neoplasms blood, Lung Neoplasms genetics, Mutation genetics, Tumor Burden genetics
- Abstract
Although programmed death-ligand 1-programmed death 1 (PD-L1-PD-1) inhibitors are broadly efficacious, improved outcomes have been observed in patients with high PD-L1 expression or high tumor mutational burden (TMB). PD-L1 testing is required for checkpoint inhibitor monotherapy in front-line non-small-cell lung cancer (NSCLC). However, obtaining adequate tumor tissue for molecular testing in patients with advanced disease can be challenging. Thus, an unmet medical need exists for diagnostic approaches that do not require tissue to identify patients who may benefit from immunotherapy. Here, we describe a novel, technically robust, blood-based assay to measure TMB in plasma (bTMB) that is distinct from tissue-based approaches. Using a retrospective analysis of two large randomized trials as test and validation studies, we show that bTMB reproducibly identifies patients who derive clinically significant improvements in progression-free survival from atezolizumab (an anti-PD-L1) in second-line and higher NSCLC. Collectively, our data show that high bTMB is a clinically actionable biomarker for atezolizumab in NSCLC.
- Published
- 2018
- Full Text
- View/download PDF
48. Phase I study of the anti-α5β1 monoclonal antibody MINT1526A with or without bevacizumab in patients with advanced solid tumors.
- Author
-
Weekes CD, Rosen LS, Capasso A, Wong KM, Ye W, Anderson M, McCall B, Fredrickson J, Wakshull E, Eppler S, Shon-Nguyen Q, Desai R, Huseni M, Hegde PS, Pourmohamad T, Rhee I, and Bessudo A
- Subjects
- Adult, Aged, Aged, 80 and over, Antineoplastic Agents, Immunological immunology, Antineoplastic Combined Chemotherapy Protocols immunology, Bevacizumab administration & dosage, Bevacizumab immunology, Dose-Response Relationship, Drug, Female, Humans, Integrin alpha5beta1 antagonists & inhibitors, Integrin alpha5beta1 immunology, Male, Middle Aged, Neoplasms immunology, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Neoplasms drug therapy
- Abstract
Purpose: MINT1526A is a monoclonal antibody that blocks the interaction of integrin alpha 5 beta 1 (α5β1) with its extracellular matrix ligands. This phase I study evaluated the safety and pharmacokinetics of MINT1526A with or without bevacizumab in patients with advanced solid tumors., Methods: MINT1526A was administered every 3 weeks (Q3W) as monotherapy (arm 1) or in combination with bevacizumab 15 mg/kg, Q3W (arm 2). Each arm included a 3 + 3 dose-escalation stage and a dose-expansion stage., Results: Twenty-four patients were enrolled in arm 1 (dose range 2-30 mg/kg) and 30 patients were enrolled in arm 2 (dose range 3-15 mg/kg). Monocyte α5β1 receptor occupancy was saturated at a dose of 15 mg/kg. No dose-limiting toxicities were observed, and the maximum tolerated dose was not reached in either arm. The most common adverse events, regardless of causality, included abdominal pain (25%), diarrhea (25%), nausea (21%), vomiting (21%), and fatigue (21%) in arm 1 and nausea (40%), fatigue (33%), vomiting (30%), dehydration (30%), headache (30%), and hypertension (30%) in arm 2. No grade ≥ 3 bleeding events were observed in either arm. No confirmed partial responses (PR) were observed in arm 1. In arm 2, one patient with thymic carcinoma experienced a confirmed PR and two patients with hepatocellular carcinoma (HCC) experienced durable minor radiographic responses., Conclusions: MINT1526A, with or without bevacizumab, was well-tolerated. Preliminary evidence of combination efficacy, including in patients with HCC, was observed, but cannot be distinguished from bevacizumab monotherapy in this phase I study.
- Published
- 2018
- Full Text
- View/download PDF
49. Interrogation of transcriptomic changes associated with drug-induced hepatic sinusoidal dilatation in colorectal cancer.
- Author
-
Jarzabek MA, Proctor WR, Vogt J, Desai R, Dicker P, Cain G, Raja R, Brodbeck J, Stevens D, van der Stok EP, Martens JWM, Verhoef C, Hegde PS, Byrne AT, and Tarrant JM
- Subjects
- Aged, Animals, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biopsy, Capillaries drug effects, Capillaries metabolism, Capillaries pathology, Colorectal Neoplasms pathology, Dilatation, Pathologic chemically induced, Dilatation, Pathologic genetics, Female, Gene Expression Profiling, Humans, Liver blood supply, Liver metabolism, Liver Neoplasms secondary, Macaca fascicularis, Male, Middle Aged, Neovascularization, Pathologic chemically induced, Neovascularization, Pathologic genetics, Oxaliplatin administration & dosage, Chemical and Drug Induced Liver Injury genetics, Colorectal Neoplasms drug therapy, Liver drug effects, Liver pathology, Oxaliplatin adverse effects, Transcriptome drug effects
- Abstract
Drug-related sinusoidal dilatation (SD) is a common form of hepatotoxicity associated with oxaliplatin-based chemotherapy used prior to resection of colorectal liver metastases (CRLM). Recently, hepatic SD has also been associated with anti-delta like 4 (DLL4) cancer therapies targeting the NOTCH pathway. To investigate the hypothesis that NOTCH signaling plays an important role in drug-induced SD, gene expression changes were examined in livers from anti-DLL4 and oxaliplatin-induced SD in non-human primate (NHP) and patients, respectively. Putative mechanistic biomarkers of bevacizumab (bev)-mediated protection against oxaliplatin-induced SD were also investigated. RNA was extracted from whole liver sections or centrilobular regions by laser-capture microdissection (LCM) obtained from NHP administered anti-DLL4 fragment antigen-binding (F(ab')2 or patients with CRLM receiving oxaliplatin-based chemotherapy with or without bev. mRNA expression was quantified using high-throughput real-time quantitative PCR. Significance analysis was used to identify genes with differential expression patterns (false discovery rate (FDR) < 0.05). Eleven (CCL2, CCND1, EFNB2, ERG, ICAM1, IL16, LFNG, NOTCH1, NOTCH4, PRDX1, and TGFB1) and six (CDH5, EFNB2, HES1, IL16, MIK67, HES1 and VWF) candidate genes were differentially expressed in the liver of anti-DLL4- and oxaliplatin-induced SD, respectively. Addition of bev to oxaliplatin-based chemotherapy resulted in differential changes in hepatic CDH5, HEY1, IL16, JAG1, MMP9, NOTCH4 and TIMP1 expression. This work implicates NOTCH and IL16 pathways in the pathogenesis of drug-induced SD and further explains the hepato-protective effect of bev in oxaliplatin-induced SD observed in CRLM patients., Competing Interests: We have the following interests. This study was partly funded by Genentech, Inc. WRP, JV, RD, GC, RR, JB, DS, PSH, and JMT were employed by Genentech, Inc. during their contributions to this work. RR is now a paid employee of MedImmune AstraZeneca. JB is now a paid employee of Merck. JMT is now a paid employee of Gilead. Genentech paid for the NHP study. There are no patents, products in development or marketed products to declare. This does not alter our adherence to all the PLOS ONE policies on sharing data and materials as detailed in the guide for authors.
- Published
- 2018
- Full Text
- View/download PDF
50. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma.
- Author
-
McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, Fong L, Joseph RW, Pal SK, Reeves JA, Sznol M, Hainsworth J, Rathmell WK, Stadler WM, Hutson T, Gore ME, Ravaud A, Bracarda S, Suárez C, Danielli R, Gruenwald V, Choueiri TK, Nickles D, Jhunjhunwala S, Piault-Louis E, Thobhani A, Qiu J, Chen DS, Hegde PS, Schiff C, Fine GD, and Powles T
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacology, Antibodies, Monoclonal, Humanized, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols pharmacology, Bevacizumab adverse effects, Bevacizumab pharmacology, Female, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Mutation genetics, Sunitinib adverse effects, Sunitinib pharmacology, Treatment Outcome, Antibodies, Monoclonal therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bevacizumab therapeutic use, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell genetics, Kidney Neoplasms drug therapy, Kidney Neoplasms genetics, Sunitinib therapeutic use
- Abstract
We describe results from IMmotion150, a randomized phase 2 study of atezolizumab (anti-PD-L1) alone or combined with bevacizumab (anti-VEGF) versus sunitinib in 305 patients with treatment-naive metastatic renal cell carcinoma. Co-primary endpoints were progression-free survival (PFS) in intent-to-treat and PD-L1+ populations. Intent-to-treat PFS hazard ratios for atezolizumab + bevacizumab or atezolizumab monotherapy versus sunitinib were 1.0 (95% confidence interval (CI), 0.69-1.45) and 1.19 (95% CI, 0.82-1.71), respectively; PD-L1+ PFS hazard ratios were 0.64 (95% CI, 0.38-1.08) and 1.03 (95% CI, 0.63-1.67), respectively. Exploratory biomarker analyses indicated that tumor mutation and neoantigen burden were not associated with PFS. Angiogenesis, T-effector/IFN-γ response, and myeloid inflammatory gene expression signatures were strongly and differentially associated with PFS within and across the treatments. These molecular profiles suggest that prediction of outcomes with anti-VEGF and immunotherapy may be possible and offer mechanistic insights into how blocking VEGF may overcome resistance to immune checkpoint blockade.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.