167 results on '"Jeffrey M. Conroy"'
Search Results
2. Case report: Single gene testing and comprehensive genomic profiling in non-small cell lung cancer—a case series of divergent results from a large reference laboratory
- Author
-
Kyle C. Strickland, Mary K. Nesline, Rebecca A. Previs, Heidi Ko, Maureen Cooper, Jennifer R. Rushton, Zachary D. Wallen, Sarabjot Pabla, Jeffrey M. Conroy, Mark Sausen, Kamal S. Saini, Luca Cantini, Taylor J. Jensen, Brian J. Caveney, Marcia Eisenberg, Eric A. Severson, and Shakti Ramkissoon
- Subjects
non-small cell lung cancer (NSCLC) molecular testing ,single gene testing (SGT) ,comprehensive genomic profiling (CGP) ,next-generation sequencing (NGS) ,cancer genomics ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Clinical management of non-small cell lung cancer (NSCLC) requires accurate identification of tumor-specific genetic alterations to inform treatment options. Historically, providers have relied on single-gene testing (SGT) for actionable variants due to a perception of cost-effectiveness and/or efficient turnaround time compared to next-generation sequencing (NGS). However, not all actionable variants may be evaluated through SGT modalities, and an SGT approach can exhaust valuable tissue needed for more comprehensive analyses. In contrast, comprehensive genomic profiling (CGP) tests employ NGS to sequence megabases of DNA and RNA to evaluate all relevant molecular alterations, providing a broader genetic profile to identify actionable alterations that SGT may not accurately or efficiently assess. Here, we briefly describe four cases from a large reference laboratory in which actionable alterations were identified by CGP but not SGT. The discussion highlights the utility and advantages of using CGP to provide complete and timely treatment options and clinical trial opportunities for patients with NSCLC.
- Published
- 2024
- Full Text
- View/download PDF
3. Cancer testis antigen burden (CTAB): a novel biomarker of tumor-associated antigens in lung cancer
- Author
-
R. J. Seager, Maria-Fernanda Senosain, Erik Van Roey, Shuang Gao, Paul DePietro, Mary K. Nesline, Durga Prasad Dash, Shengle Zhang, Heidi Ko, Stephanie B. Hastings, Kyle C. Strickland, Rebecca A. Previs, Taylor J. Jensen, Marcia Eisenberg, Brian J. Caveney, Eric A. Severson, Shakti Ramkissoon, Jeffrey M. Conroy, and Sarabjot Pabla
- Subjects
Tumor microenvironment ,Inflammation ,Immunotherapy ,Immune checkpoint inhibitors ,Gene expression profiling ,Medicine - Abstract
Abstract Background Cancer-testis antigens (CTAs) are tumor antigens that are normally expressed in the testes but are aberrantly expressed in several cancers. CTA overexpression drives the metastasis and progression of lung cancer, and is associated with poor prognosis. To improve lung cancer diagnosis, prognostic prediction, and drug discovery, robust CTA identification and quantitation is needed. In this study, we examined and quantified the co-expression of CTAs in lung cancer to derive cancer testis antigen burden (CTAB), a novel biomarker of immunotherapy response. Methods Formalin fixed paraffin embedded (FFPE) tumor samples in discovery cohort (n = 5250) and immunotherapy and combination therapy treated non-small cell lung cancer (NSCLC) retrospective (n = 250) cohorts were tested by comprehensive genomic and immune profiling (CGIP), including tumor mutational burden (TMB) and the mRNA expression of 17 CTAs. PD-L1 expression was evaluated by IHC. CTA expression was summed to derive the CTAB score. The median CTAB score for the discovery cohort of 170 was applied to the retrospective cohort as cutoff for CTAB “high” and “low”. Biomarker and gene expression correlation was measured by Spearman correlation. Kaplan–Meier survival analyses were used to detect overall survival (OS) differences, and objective response rate (ORR) based on RECIST criteria was compared using Fisher’s exact test. Results The CTAs were highly co-expressed (p
- Published
- 2024
- Full Text
- View/download PDF
4. High indoleamine 2,3-dioxygenase transcript levels predict better outcome after front-line cancer immunotherapy
- Author
-
Yu Fujiwara, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Suzanna Lee, Mary K. Nesline, Jeffrey M. Conroy, Paul DePietro, Sarabjot Pabla, Scott M. Lippman, and Razelle Kurzrock
- Subjects
Immunology ,Molecular biology ,Cancer ,Science - Abstract
Summary: Indoleamine 2,3-dioxygenase 1 (IDO1), which catabolizes tryptophan, is a potential target to unlock the immunosuppressive tumor microenvironment. Correlations between IDO1 and immune checkpoint inhibitor (ICI) efficacy remain unclear. Herein, we investigated IDO1 transcript expression across cancers and clinical outcome correlations. High IDO1 transcripts were more frequent in uterine (54.2%) and ovarian cancer (37.2%) but varied between and within malignancies. High IDO1 RNA expression was associated with high expression of PD-L1 (immune checkpoint ligand), CXCL10 (an effector T cell recruitment chemokine), and STAT1 (a component of the JAK-STAT pathway) (all multivariable p
- Published
- 2024
- Full Text
- View/download PDF
5. T-cell priming transcriptomic markers: implications of immunome heterogeneity for precision immunotherapy
- Author
-
Hirotaka Miyashita, Razelle Kurzrock, Nicholas J. Bevins, Kartheeswaran Thangathurai, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Sean T. Glenn, Jeffrey M. Conroy, Paul DePietro, Eitan Rubin, Jason K. Sicklick, and Shumei Kato
- Subjects
Medicine ,Genetics ,QH426-470 - Abstract
Abstract Immune checkpoint blockade is effective for only a subset of cancers. Targeting T-cell priming markers (TPMs) may enhance activity, but proper application of these agents in the clinic is challenging due to immune complexity and heterogeneity. We interrogated transcriptomics of 15 TPMs (CD137, CD27, CD28, CD80, CD86, CD40, CD40LG, GITR, ICOS, ICOSLG, OX40, OX40LG, GZMB, IFNG, and TBX21) in a pan-cancer cohort (N = 514 patients, 30 types of cancer). TPM expression was analyzed for correlation with histological type, microsatellite instability high (MSI-H), tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) expression. Among 514 patients, the most common histological types were colorectal (27%), pancreatic (11%), and breast cancer (10%). No statistically significant association between histological type and TPM expression was seen. In contrast, expression of GZMB (granzyme B, a serine protease stored in activated T and NK cells that induces cancer cell apoptosis) and IFNG (activates cytotoxic T cells) were significantly higher in tumors with MSI-H, TMB ≥ 10 mutations/mb and PD-L1 ≥ 1%. PD-L1 ≥ 1% was also associated with significantly higher CD137, GITR, and ICOS expression. Patients’ tumors were classified into “Hot”, “Mixed”, or “Cold” clusters based on TPM expression using hierarchical clustering. The cold cluster showed a significantly lower proportion of tumors with PD-L1 ≥ 1%. Overall, 502 patients (98%) had individually distinct patterns of TPM expression. Diverse expression patterns of TPMs independent of histological type but correlating with other immunotherapy biomarkers (PD-L1 ≥ 1%, MSI-H and TMB ≥ 10 mutations/mb) were observed. Individualized selection of patients based on TPM immunomic profiles may potentially help with immunotherapy optimization.
- Published
- 2023
- Full Text
- View/download PDF
6. High CTLA-4 transcriptomic expression correlates with high expression of other checkpoints and with immunotherapy outcome
- Author
-
Nithya Krishnamurthy, Daisuke Nishizaki, Scott M. Lippman, Hirotaka Miyashita, Mary K. Nesline, Sarabjot Pabla, Jeffrey M. Conroy, Paul DePietro, Shumei Kato, and Razelle Kurzrock
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: CTLA-4 impedes the immune system’s antitumor response. There are two Food and Drug Administration-approved anti-CTLA-4 agents – ipilimumab and tremelimumab – both used together with anti-PD-1/PD-L1 agents. Objective: To assess the prognostic implications and immunologic correlates of high CTLA-4 in tumors of patients on immunotherapy and those on non-immunotherapy treatments. Design/methods: We evaluated RNA expression levels in a clinical-grade laboratory and clinical correlates of CTLA-4 and other immune checkpoints in 514 tumors, including 489 patients with advanced/metastatic cancers and full outcome annotation. A reference population (735 tumors; 35 histologies) was used to normalize and rank transcript abundance (0–100 percentile) to internal housekeeping gene profiles. Results: The most common tumor types were colorectal (140/514, 27%), pancreatic (55/514, 11%), breast (49/514, 10%), and ovarian cancers (43/514, 8%). Overall, 87 of 514 tumors (16.9%) had high CTLA-4 transcript expression (⩾75th percentile rank). Cancers with the largest proportion of high CTLA-4 transcripts were cervical cancer (80% of patients), small intestine cancer (33.3%), and melanoma (33.3%). High CTLA-4 RNA independently/significantly correlated with high PD-1, PD- L2, and LAG3 RNA levels (and with high PD-L1 in univariate analysis). High CTLA-4 RNA expression was not correlated with survival from the time of metastatic disease [ N = 272 patients who never received immune checkpoint inhibitors (ICIs)]. However, in 217 patients treated with ICIs (mostly anti-PD-1/anti-PD- L1), progression-free survival (PFS) and overall survival (OS) were significantly longer among patients with high versus non-high CTLA-4 expression [hazard ratio, 95% confidence interval: 0.6 (0.4–0.9) p = 0.008; and 0.5 (0.3–0.8) p = 0.002, respectively]; results were unchanged when 18 patients who received anti-CTLA-4 were omitted. Patients whose tumors had high CTLA-4 and high PD-L1 did best; those with high PD-L1 but non-high CTLA-4 and/or other expression patterns had poorer outcomes for PFS ( p = 0.004) and OS ( p = 0.009) after immunotherapy. Conclusion: High CTLA-4, especially when combined with high PD-L1 transcript expression, was a significant positive predictive biomarker for better outcomes (PFS and OS) in patients on immunotherapy.
- Published
- 2024
- Full Text
- View/download PDF
7. Pan‐cancer analysis of TIM‐3 transcriptomic expression reveals high levels in pancreatic cancer and interpatient heterogeneity
- Author
-
Jungah Lim, Razelle Kurzrock, Daisuke Nishizaki, Hirotaka Miyashita, Jacob J. Adashek, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Jeffrey M. Conroy, Paul DePietro, Scott M. Lippman, and Shumei Kato
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background T‐cell immunoglobulin and mucin domain‐containing protein 3 (TIM‐3), an immune checkpoint receptor, dampens immune function. TIM‐3 antagonists have entered the clinic. Methods We analyzed TIM‐3 transcriptomic expression in 514 diverse cancers. Transcript abundance was normalized to internal housekeeping genes and ranked (0–100 percentile) to a reference population (735 tumors; 35 histologies [high≥75 percentile rank]). Ninety tumors (17.5%) demonstrated high TIM‐3 expression. Results TIM‐3 expression varied between and within tumor types. However, high TIM‐3 expression was more common in pancreatic cancer (20/55 tumors, 36.4%; odds ratio, 95% confidence interval (pancreatic vs. other tumors) = 3.176 (1.733–5.818; p
- Published
- 2024
- Full Text
- View/download PDF
8. LAG‐3 transcriptomic expression patterns across malignancies: Implications for precision immunotherapeutics
- Author
-
Jacob J. Adashek, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Pradip De, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Jeffrey M. Conroy, Paul DePietro, Scott Lippman, and Razelle Kurzrock
- Subjects
biomarkers ,clinical trials ,experimental therapeutics ,immune checkpoints ,immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Lymphocyte activation gene 3 (LAG‐3) or CD223 is a transmembrane protein that serves as an immune checkpoint which attenuates T‐cell activation. Many clinical trials of LAG‐3 inhibitors have had modest effects, but recent data indicate that the LAG‐3 antibody relatlimab, together with nivolumab (anti‐PD‐1), provided greater benefit than nivolumab alone in patients with melanoma. Methods In this study, the RNA expression levels of 397 genes were assessed in 514 diverse cancers at a clinical‐grade laboratory (OmniSeq: https://www.omniseq.com/). Transcript abundance was normalized to internal housekeeping gene profiles and ranked (0–100 percentile) using a reference population (735 tumors; 35 histologies). Results A total of 116 of 514 tumors (22.6%) had high LAG‐3 transcript expression (≥75 percentile rank). Cancers with the greatest proportion of high LAG‐3 transcripts were neuroendocrine (47% of patients) and uterine (42%); colorectal had among the lowest proportion of high LAG‐3 expression (15% of patients) (all p
- Published
- 2023
- Full Text
- View/download PDF
9. Integration of tumor inflammation, cell proliferation, and traditional biomarkers improves prediction of immunotherapy resistance and response
- Author
-
Sarabjot Pabla, R. J. Seager, Erik Van Roey, Shuang Gao, Carrie Hoefer, Mary K. Nesline, Paul DePietro, Blake Burgher, Jonathan Andreas, Vincent Giamo, Yirong Wang, Felicia L. Lenzo, Margot Schoenborn, Shengle Zhang, Roger Klein, Sean T. Glenn, and Jeffrey M. Conroy
- Subjects
Inflammation ,Cell proliferation ,Pembrolizumab ,Nivolumab ,Ipilimumab ,Algorithmic analysis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background Contemporary to the rapidly evolving landscape of cancer immunotherapy is the equally changing understanding of immune tumor microenvironments (TMEs) which is crucial to the success of these therapies. Their reliance on a robust host immune response necessitates clinical grade measurements of immune TMEs at diagnosis. In this study, we describe a stable tumor immunogenic profile describing immune TMEs in multiple tumor types with ability to predict clinical benefit from immune checkpoint inhibitors (ICIs). Methods A tumor immunogenic signature (TIGS) was derived from targeted RNA-sequencing (RNA-seq) and gene expression analysis of 1323 clinical solid tumor cases spanning 35 histologies using unsupervised analysis. TIGS correlation with ICI response and survival was assessed in a retrospective cohort of NSCLC, melanoma and RCC tumor blocks, alone and combined with TMB, PD-L1 IHC and cell proliferation biomarkers. Results Unsupervised clustering of RNA-seq profiles uncovered a 161 gene signature where T cell and B cell activation, IFNg, chemokine, cytokine and interleukin pathways are over-represented. Mean expression of these genes produced three distinct TIGS score categories: strong (n = 384/1323; 29.02%), moderate (n = 354/1323; 26.76%), and weak (n = 585/1323; 44.22%). Strong TIGS tumors presented an improved ICI response rate of 37% (30/81); with highest response rate advantage occurring in NSCLC (ORR = 36.6%; 16/44; p = 0.051). Similarly, overall survival for strong TIGS tumors trended upward (median = 25 months; p = 0.19). Integrating the TIGS score categories with neoplastic influence quantified via cell proliferation showed highly proliferative and strong TIGS tumors correlate with significantly higher ICI ORR than poorly proliferative and weak TIGS tumors [14.28%; p = 0.0006]. Importantly, we noted that strong TIGS and highly [median = not achieved; p = 0.025] or moderately [median = 16.2 months; p = 0.025] proliferative tumors had significantly better survival compared to weak TIGS, highly proliferative tumors [median = 7.03 months]. Importantly, TIGS discriminates subpopulations of potential ICI responders that were considered negative for response by TMB and PD-L1. Conclusions TIGS is a comprehensive and informative measurement of immune TME that effectively characterizes host immune response to ICIs in multiple tumors. The results indicate that when combined with PD-L1, TMB and cell proliferation, TIGS provides greater context of both immune and neoplastic influences on the TME for implementation into clinical practice.
- Published
- 2021
- Full Text
- View/download PDF
10. Proliferative potential and resistance to immune checkpoint blockade in lung cancer patients
- Author
-
Sarabjot Pabla, Jeffrey M. Conroy, Mary K. Nesline, Sean T. Glenn, Antonios Papanicolau-Sengos, Blake Burgher, Jacob Hagen, Vincent Giamo, Jonathan Andreas, Felicia L. Lenzo, Wang Yirong, Grace K. Dy, Edwin Yau, Amy Early, Hongbin Chen, Wiam Bshara, Katherine G. Madden, Keisuke Shirai, Konstantin Dragnev, Laura J. Tafe, Daniele Marin, Jason Zhu, Jeff Clarke, Matthew Labriola, Shannon McCall, Tian Zhang, Matthew Zibelman, Pooja Ghatalia, Isabel Araujo-Fernandez, Arun Singavi, Ben George, Andrew Craig MacKinnon, Jonathan Thompson, Rajbir Singh, Robin Jacob, Lynn Dressler, Mark Steciuk, Oliver Binns, Deepa Kasuganti, Neel Shah, Marc Ernstoff, Kunle Odunsi, Razelle Kurzrock, Mark Gardner, Lorenzo Galluzzi, and Carl Morrison
- Subjects
Atezolizumab ,Nivolumab ,Pembrolizumab ,Ipilimumab ,PD-1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Resistance to immune checkpoint inhibitors (ICIs) has been linked to local immunosuppression independent of major ICI targets (e.g., PD-1). Clinical experience with response prediction based on PD-L1 expression suggests that other factors influence sensitivity to ICIs in non-small cell lung cancer (NSCLC) patients. Methods Tumor specimens from 120 NSCLC patients from 10 institutions were evaluated for PD-L1 expression by immunohistochemistry, and global proliferative profile by targeted RNA-seq. Results Cell proliferation, derived from the mean expression of 10 proliferation-associated genes (namely BUB1, CCNB2, CDK1, CDKN3, FOXM1, KIAA0101, MAD2L1, MELK, MKI67, and TOP2A), was identified as a marker of response to ICIs in NSCLC. Poorly, moderately, and highly proliferative tumors were somewhat equally represented in NSCLC, with tumors with the highest PD-L1 expression being more frequently moderately proliferative as compared to lesser levels of PD-L1 expression. Proliferation status had an impact on survival in patients with both PD-L1 positive and negative tumors. There was a significant survival advantage for moderately proliferative tumors compared to their combined highly/poorly counterparts (p = 0.021). Moderately proliferative PD-L1 positive tumors had a median survival of 14.6 months that was almost twice that of PD-L1 negative highly/poorly proliferative at 7.6 months (p = 0.028). Median survival in moderately proliferative PD-L1 negative tumors at 12.6 months was comparable to that of highly/poorly proliferative PD-L1 positive tumors at 11.5 months, but in both instances less than that of moderately proliferative PD-L1 positive tumors. Similar to survival, proliferation status has impact on disease control (DC) in patients with both PD-L1 positive and negative tumors. Patients with moderately versus those with poorly or highly proliferative tumors have a superior DC rate when combined with any classification schema used to score PD-L1 as a positive result (i.e., TPS ≥ 50% or ≥ 1%), and best displayed by a DC rate for moderately proliferative tumors of no less than 40% for any classification of PD-L1 as a negative result. While there is an over representation of moderately proliferative tumors as PD-L1 expression increases this does not account for the improved survival or higher disease control rates seen in PD-L1 negative tumors. Conclusions Cell proliferation is potentially a new biomarker of response to ICIs in NSCLC and is applicable to PD-L1 negative tumors.
- Published
- 2019
- Full Text
- View/download PDF
11. Treatment recommendations to cancer patients in the context of FDA guidance for next generation sequencing
- Author
-
Grace K. Dy, Mary K. Nesline, Antonios Papanicolau-Sengos, Paul DePietro, Charles M. LeVea, Amy Early, Hongbin Chen, Anne Grand’Maison, Patrick Boland, Marc S. Ernstoff, Stephen Edge, Stacey Akers, Mateusz Opyrchal, Gurkamal Chatta, Kunle Odunsi, Sarabjot Pabla, Jeffrey M. Conroy, Sean T. Glenn, Hanchun T. DeFedericis, Blake Burgher, Jonathan Andreas, Vincent Giamo, Maochun Qin, Yirong Wang, Kazunori Kanehira, Felicia L. Lenzo, Peter Frederick, Shashikant Lele, Lorenzo Galluzzi, Boris Kuvshinoff, and Carl Morrison
- Subjects
Next generation sequencing ,Comprehensive genomic profiling ,FDA guidance ,Physician treatment recommendations ,Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
Abstract Background Regulatory approval of next generation sequencing (NGS) by the FDA is advancing the use of genomic-based precision medicine for the therapeutic management of cancer as standard care. Recent FDA guidance for the classification of genomic variants based on clinical evidence to aid clinicians in understanding the actionability of identified variants provided by comprehensive NGS panels has also been set forth. In this retrospective analysis, we interpreted and applied the FDA variant classification guidance to comprehensive NGS testing performed for advanced cancer patients and assessed oncologist agreement with NGS test treatment recommendations. Methods NGS comprehensive genomic profiling was performed in a CLIA certified lab (657 completed tests for 646 patients treated at Roswell Park Comprehensive Cancer Center) between June 2016 and June 2017. Physician treatment recommendations made within 120 days post-test were gathered from tested patients’ medical records and classified as targeted therapy, precision medicine clinical trial, immunotherapy, hormonal therapy, chemotherapy/radiation, surgery, transplant, or non-therapeutic (hospice, surveillance, or palliative care). Agreement between NGS test report targeted therapy recommendations based on the FDA variant classification and physician targeted therapy treatment recommendations were evaluated. Results Excluding variants contraindicating targeted therapy (i.e., KRAS or NRAS mutations), at least one variant with FDA level 1 companion diagnostic supporting evidence as the most actionable was identified in 14% of tests, with physicians most frequently recommending targeted therapy (48%) for patients with these results. This stands in contrast to physicians recommending targeted therapy based on test results with FDA level 2 (practice guideline) or FDA level 3 (clinical trial or off label) evidence as the most actionable result (11 and 4%, respectively). Conclusions We found an appropriate “dose-response” relationship between the strength of clinical evidence supporting biomarker-directed targeted therapy based on application of FDA guidance for NGS test variant classification, and subsequent treatment recommendations made by treating physicians. In view of recent changes at FDA, it is paramount to define regulatory grounds and medical policy coverage for NGS testing based on this guidance.
- Published
- 2019
- Full Text
- View/download PDF
12. Next generation sequencing of PD-L1 for predicting response to immune checkpoint inhibitors
- Author
-
Jeffrey M. Conroy, Sarabjot Pabla, Mary K. Nesline, Sean T. Glenn, Antonios Papanicolau-Sengos, Blake Burgher, Jonathan Andreas, Vincent Giamo, Yirong Wang, Felicia L. Lenzo, Wiam Bshara, Maya Khalil, Grace K. Dy, Katherine G. Madden, Keisuke Shirai, Konstantin Dragnev, Laura J. Tafe, Jason Zhu, Matthew Labriola, Daniele Marin, Shannon J. McCall, Jeffrey Clarke, Daniel J. George, Tian Zhang, Matthew Zibelman, Pooja Ghatalia, Isabel Araujo-Fernandez, Luis de la Cruz-Merino, Arun Singavi, Ben George, Alexander C. MacKinnon, Jonathan Thompson, Rajbir Singh, Robin Jacob, Deepa Kasuganti, Neel Shah, Roger Day, Lorenzo Galluzzi, Mark Gardner, and Carl Morrison
- Subjects
Atezolizumab ,Avelumab ,cancer immunotherapy ,Durvalumab ,Nivolumab ,Pembrolizumab ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background PD-L1 immunohistochemistry (IHC) has been traditionally used for predicting clinical responses to immune checkpoint inhibitors (ICIs). However, there are at least 4 different assays and antibodies used for PD-L1 IHC, each developed with a different ICI. We set to test if next generation RNA sequencing (RNA-seq) is a robust method to determine PD-L1 mRNA expression levels and furthermore, efficacy of predicting response to ICIs as compared to routinely used, standardized IHC procedures. Methods A total of 209 cancer patients treated on-label by FDA-approved ICIs, with evaluable responses were assessed for PD-L1 expression by RNA-seq and IHC, based on tumor proportion score (TPS) and immune cell staining (ICS). A subset of serially diluted cases was evaluated for RNA-seq assay performance across a broad range of PD-L1 expression levels. Results Assessment of PD-L1 mRNA levels by RNA-seq demonstrated robust linearity across high and low expression ranges. PD-L1 mRNA levels assessed by RNA-seq and IHC (TPS and ICS) were highly correlated (p
- Published
- 2019
- Full Text
- View/download PDF
13. Proliferative potential and response to nivolumab in clear cell renal cell carcinoma patients
- Author
-
Tian Zhang, Sarabjot Pabla, Felicia L. Lenzo, Jeffrey M. Conroy, Mary K. Nesline, Sean T. Glenn, Antonios Papanicolau-Sengos, Blake Burgher, Vincent Giamo, Jonathan Andreas, Yirong Wang, Wiam Bshara, Katherine G. Madden, Keisuke Shirai, Konstantin Dragnev, Laura J. Tafe, Rajan Gupta, Jason Zhu, Matthew Labriola, Shannon McCall, Daniel J. George, Pooja Ghatalia, Farshid Dayyani, Robert Edwards, Michelle S Park, Rajbir Singh, Robin Jacob, Saby George, Bo Xu, Matthew Zibelman, Razelle Kurzrock, and Carl Morrison
- Subjects
nivolumab ,renal cell carcinoma ,pd-1 ,pd-l1 ,proliferation ,ki-67 ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Biomarkers predicting immunotherapy response in metastatic renal cell cancer (mRCC) are lacking. PD-L1 immunohistochemistry is a complementary diagnostic for immune checkpoint inhibitors (ICIs) in mRCC, but has shown minimal clinical utility and is not used in routine clinical practice. Methods Tumor specimens from 56 patients with mRCC who received nivolumab were evaluated for PD-L1, cell proliferation (targeted RNA-seq), and outcome. Results For 56 patients treated with nivolumab as a standard of care, there were 2 complete responses and 8 partial responses for a response rate of 17.9%. Dividing cell proliferation into tertiles, derived from the mean expression of 10 proliferation-associated genes in a reference set of tumors, poorly proliferative tumors (62.5%) were more common than moderately (30.4%) or highly proliferative (8.9%) counterparts. Moderately proliferative tumors were enriched for PD-L1 positive (41.2%), compared to poorly proliferative counterparts (11.4%). Objective response for moderately proliferative (29.4%) tumors was higher than that of poorly (11.4%) proliferative counterparts, but not statistically significant (p = .11). When cell proliferation and negative PD-L1 tumor proportion scores were combined statistically significant results were achieved (p = .048), showing that patients with poorly proliferative and PD-L1 negative tumors have a very low response rate (6.5%) compared to moderately proliferative PD-L1 negative tumors (30%). Conclusions Cell proliferation has value in predicting response to nivolumab in clear cell mRCC patients, especially when combined with PD-L1 expression. Further studies which include the addition of progression-free survival (PFS) along with sufficiently powered subgroups are required to further support these findings.
- Published
- 2020
- Full Text
- View/download PDF
14. Predicting response to checkpoint inhibitors in melanoma beyond PD-L1 and mutational burden
- Author
-
Carl Morrison, Sarabjot Pabla, Jeffrey M. Conroy, Mary K. Nesline, Sean T. Glenn, Devin Dressman, Antonios Papanicolau-Sengos, Blake Burgher, Jonathan Andreas, Vincent Giamo, Moachun Qin, Yirong Wang, Felicia L. Lenzo, Angela Omilian, Wiam Bshara, Matthew Zibelman, Pooja Ghatalia, Konstantin Dragnev, Keisuke Shirai, Katherine G. Madden, Laura J. Tafe, Neel Shah, Deepa Kasuganti, Luis de la Cruz-Merino, Isabel Araujo, Yvonne Saenger, Margaret Bogardus, Miguel Villalona-Calero, Zuanel Diaz, Roger Day, Marcia Eisenberg, Steven M. Anderson, Igor Puzanov, Lorenzo Galluzzi, Mark Gardner, and Marc S. Ernstoff
- Subjects
Pembrolizumab ,Nivolumab ,Ipilimumab ,Algorithmic analysis ,Inflamed ,Borderline ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Immune checkpoint inhibitors (ICIs) have changed the clinical management of melanoma. However, not all patients respond, and current biomarkers including PD-L1 and mutational burden show incomplete predictive performance. The clinical validity and utility of complex biomarkers have not been studied in melanoma. Methods Cutaneous metastatic melanoma patients at eight institutions were evaluated for PD-L1 expression, CD8+ T-cell infiltration pattern, mutational burden, and 394 immune transcript expression. PD-L1 IHC and mutational burden were assessed for association with overall survival (OS) in 94 patients treated prior to ICI approval by the FDA (historical-controls), and in 137 patients treated with ICIs. Unsupervised analysis revealed distinct immune-clusters with separate response rates. This comprehensive immune profiling data were then integrated to generate a continuous Response Score (RS) based upon response criteria (RECIST v.1.1). RS was developed using a single institution training cohort (n = 48) and subsequently tested in a separate eight institution validation cohort (n = 29) to mimic a real-world clinical scenario. Results PD-L1 positivity ≥1% correlated with response and OS in ICI-treated patients, but demonstrated limited predictive performance. High mutational burden was associated with response in ICI-treated patients, but not with OS. Comprehensive immune profiling using RS demonstrated higher sensitivity (72.2%) compared to PD-L1 IHC (34.25%) and tumor mutational burden (32.5%), but with similar specificity. Conclusions In this study, the response score derived from comprehensive immune profiling in a limited melanoma cohort showed improved predictive performance as compared to PD-L1 IHC and tumor mutational burden.
- Published
- 2018
- Full Text
- View/download PDF
15. Pitfalls of improperly procured adjacent non-neoplastic tissue for somatic mutation analysis using next-generation sequencing
- Author
-
Lei Wei, Antonios Papanicolau-Sengos, Song Liu, Jianmin Wang, Jeffrey M. Conroy, Sean T. Glenn, Elizabeth Brese, Qiang Hu, Kiersten Marie Miles, Blake Burgher, Maochun Qin, Karen Head, Angela R. Omilian, Wiam Bshara, John Krolewski, Donald L. Trump, Candace S. Johnson, and Carl D. Morrison
- Subjects
Somatic mutations ,Tumor contamination ,Adjacent normal tissues ,Internal medicine ,RC31-1245 ,Genetics ,QH426-470 - Abstract
Abstract Background The rapid adoption of next-generation sequencing provides an efficient system for detecting somatic alterations in neoplasms. The detection of such alterations requires a matched non-neoplastic sample for adequate filtering of non-somatic events such as germline polymorphisms. Non-neoplastic tissue adjacent to the excised neoplasm is often used for this purpose as it is simultaneously collected and generally contains the same tissue type as the neoplasm. Following NGS analysis, we and others have frequently observed low-level somatic mutations in these non-neoplastic tissues, which may impose additional challenges to somatic mutation detection as it complicates germline variant filtering. Methods We hypothesized that the low-level somatic mutation observed in non-neoplastic tissues may be entirely or partially caused by inadvertent contamination by neoplastic cells during the surgical pathology gross assessment or tissue procurement process. To test this hypothesis, we applied a systematic protocol designed to collect multiple grossly non-neoplastic tissues using different methods surrounding each single neoplasm. The procedure was applied in two breast cancer lumpectomy specimens. In each case, all samples were first sequenced by whole-exome sequencing to identify somatic mutations in the neoplasm and determine their presence in the adjacent non-neoplastic tissues. We then generated ultra-deep coverage using targeted sequencing to assess the levels of contamination in non-neoplastic tissue samples collected under different conditions. Results Contamination levels in non-neoplastic tissues ranged up to 3.5 and 20.9 % respectively in the two cases tested, with consistent pattern correlated with the manner of grossing and procurement. By carefully controlling the conditions of various steps during this process, we were able to eliminate any detectable contamination in both patients. Conclusion The results demonstrated that the process of tissue procurement contributes to the level of contamination in non-neoplastic tissue, and contamination can be reduced to below detectable levels by using a carefully designed collection method. A standard protocol dedicated for acquiring adjacent non-neoplastic tissue that minimizes neoplasm contamination should be implemented for all somatic mutation detection studies.
- Published
- 2016
- Full Text
- View/download PDF
16. PD-L2 amplification and durable disease stabilization in patient with urothelial carcinoma receiving pembrolizumab
- Author
-
Saby George, Antonios Papanicolau-Sengos, Felicia L. Lenzo, Jeffrey M. Conroy, Mary Nesline, Sarabjot Pabla, Sean T. Glenn, Blake Burgher, Jonathan Andreas, Vincent Giamo, Moachun Qin, Yirong Wang, Lorenzo Galluzzi, and Carl Morrison
- Subjects
22c3 assay ,adora2a ,atezolizumab ,immunohistochemistry ,pd-1 ,sp142 assay ,Immunologic diseases. Allergy ,RC581-607 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We report the immunological profile of a patient with upper-tract urothelial carcinoma experiencing stable disease on pembrolizumab for 20 months. The tumor exhibited extensive infiltration by CD8+ cytotoxic T lymphocytes, low-to-moderate mutational burden, no PD-L1 staining by commercially available immunohistochemical assays, but amplification of CD274 (coding for PD-L1) and/or PDCD1LG2 (encoding PD-L2) by fluorescence in situ hybridization. RNA-seq revealed multiple biomarkers of an ongoing immune response and compensatory immune evasion, including moderate PD-L1 levels coupled with robust PD-L2 expression. Pending validation in additional patients, these findings suggest that PD-L2 expression levels may constitute a biomarker of response to immune checkpoint blockade in urothelial carcinoma.
- Published
- 2018
- Full Text
- View/download PDF
17. aCGHViewer: A Generic Visualization Tool For aCGH data
- Author
-
Ganesh Shankar, Michael R. Rossi, Devin E. McQuaid, Jeffrey M. Conroy, Daniel G. Gaile, John K. Cowell, Norma J. Nowak, and Ping Liang
- Subjects
array-CGH ,CNA ,gene expression ,visualization ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Array-Comparative Genomic Hybridization (aCGH) is a powerful high throughput technology for detecting chromosomal copy number aberrations (CNAs) in cancer, aiming at identifying related critical genes from the affected genomic regions. However, advancing from a dataset with thousands of tabular lines to a few candidate genes can be an onerous and time-consuming process. To expedite the aCGH data analysis process, we have developed a user-friendly aCGH data viewer (aCGHViewer) as a conduit between the aCGH data tables and a genome browser. The data from a given aCGH analysis are displayed in a genomic view comprised of individual chromosome panels which can be rapidly scanned for interesting features. A chromosome panel containing a feature of interest can be selected to launch a detail window for that single chromosome. Selecting a data point of interest in the detail window launches a query to the UCSC or NCBI genome browser to allow the user to explore the gene content in the chromosomal region. Additionally, aCGHViewer can display aCGH and expression array data concurrently to visually correlate the two. aCGHViewer is a stand alone Java visualization application that should be used in conjunction with separate statistical programs. It operates on all major computer platforms and is freely available at http://falcon.roswellpark.org/aCGHview/.
- Published
- 2006
18. A consensus-based classification workflow to determine genetically inferred ancestry from comprehensive genomic profiling of patients with solid tumors.
- Author
-
Zachary D. Wallen, Mary K. Nesline, Sarabjot Pabla, Shuang Gao, Erik Vanroey, Stephanie B. Hastings, Heidi Ko, Kyle C. Strickland, Rebecca A Previs, Shengle Zhang, Jeffrey M. Conroy, Taylor J. Jensen, Elizabeth George, Marcia Eisenberg, Brian Caveney, Pratheesh Sathyan, Shakti Ramkissoon, and Eric A Severson
- Published
- 2024
- Full Text
- View/download PDF
19. Association of Antifolate Response Signature Status and Clinical Activity of Pemetrexed-Platinum Chemotherapy in Non-Small Cell Lung Cancer - The Piedmont Study
- Author
-
Joel R. Eisner, Gregory M. Mayhew, James M. Davison, Kirk D. Beebe, Yoichiro Shibata, Yuelong Guo, Carol Farhangfar, Farhang Farhangfar, Joshua M. Uronis, Jeffrey M. Conroy, Michael V. Milburn, David Neil. Hayes, and Kathryn F. Mileham
- Subjects
Cancer Research ,Oncology - Abstract
Purpose: The Piedmont study is a prospectively designed retrospective evaluation of a new 48-gene antifolate response signature (AF-PRS) in patients with locally advanced/metastatic NS-NSCLC treated with pemetrexed-containing platinum doublet chemotherapy (PMX-PDC). The study tested the hypothesis that AF-PRS selects for patients with NS-NSCLC that preferentially respond to PMX-PDC, with a goal of providing clinical support for AF-PRS as potential diagnostic test. Experimental Design: Residual pre-treatment FFPE tumor samples and clinical data were analyzed from 105 patients treated with 1st-line (1L) PMX-PDC. 95 patients had sufficient RNA sequencing (RNAseq) data quality and clinical annotation for inclusion in the analysis. Associations between AF-PRS status and associate genes, and outcome measures including progression-free survival (PFS) and clinical response were evaluated. Results: Overall, 53% of patients were AF-PRS(+), which was associated with extended PFS, but not OS, vs. AF-PRS(-) (16.6 vs. 6.6 mo; p = 0.025). In patients who were Stage I-III patients at time of treatment, PFS was further extended in AF-PRS(+) vs. AF-PRS(-) (36.2 vs. 9.3 mo; p = 0.03). Complete response (CR) to therapy was noted in 14 of 95 patients. AF-PRS(+) preferentially selected a majority (79%) of CRs, which were evenly split between patients Stage I-III (6 of 7) and Stage IV (5 of 7) at time of treatment. Conclusions: AF-PRS identified a significant population of patients with extended PFS and/or clinical response following PMX-PDC treatment. AF-PRS may be a useful diagnostic test for patients indicated for systemic chemotherapy, especially when determining the optimal PDC regimen for locally advanced disease.
- Published
- 2023
- Full Text
- View/download PDF
20. <scp>LAG</scp> ‐3 transcriptomic expression patterns across malignancies: Implications for precision immunotherapeutics
- Author
-
Jacob J. Adashek, Shumei Kato, Daisuke Nishizaki, Hirotaka Miyashita, Pradip De, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Jeffrey M. Conroy, Paul DePietro, Scott Lippman, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2023
- Full Text
- View/download PDF
21. Dissection of a Down syndrome-associated trisomy to separate the gene dosage-dependent and -independent effects of an extra chromosome
- Author
-
Zhuo Xing, Yichen Li, Eduardo Cortes-Gomez, Xiaoling Jiang, Shuang Gao, Annie Pao, Jidong Shan, Yinghui Song, Amanda Perez, Tao Yu, Max R Highsmith, Frimpong Boadu, Jeffrey M Conroy, Prashant K Singh, Andrei V Bakin, Jianlin Cheng, Zhijun Duan, Jianmin Wang, Song Liu, Benjamin Tycko, and Y Eugene Yu
- Subjects
Genetics ,General Medicine ,Molecular Biology ,Genetics (clinical) - Abstract
As an aneuploidy, trisomy is associated with mammalian embryonic and postnatal abnormalities. Understanding the underlying mechanisms involved in mutant phenotypes is broadly important and may lead to new strategies to treat clinical manifestations in individuals with trisomies, such as trisomy 21 [Down syndrome (DS)]. Although increased gene dosage effects because of a trisomy may account for the mutant phenotypes, there is also the possibility that phenotypic consequences of a trisomy can arise because of the presence of a freely segregating extra chromosome with its own centromere, i.e. a ‘free trisomy’ independent of gene dosage effects. Presently, there are no reports of attempts to functionally separate these two types of effects in mammals. To fill this gap, here we describe a strategy that employed two new mouse models of DS, Ts65Dn;Df(17)2Yey/+ and Dp(16)1Yey/Df(16)8Yey. Both models carry triplications of the same 103 human chromosome 21 gene orthologs; however, only Ts65Dn;Df(17)2Yey/+ mice carry a free trisomy. Comparison of these models revealed the gene dosage-independent impacts of an extra chromosome at the phenotypic and molecular levels for the first time. They are reflected by impairments of Ts65Dn;Df(17)2Yey/+ males in T-maze tests when compared with Dp(16)1Yey/Df(16)8Yey males. Results from the transcriptomic analysis suggest the extra chromosome plays a major role in trisomy-associated expression alterations of disomic genes beyond gene dosage effects. This model system can now be used to deepen our mechanistic understanding of this common human aneuploidy and obtain new insights into the effects of free trisomies in other human diseases such as cancers.
- Published
- 2023
- Full Text
- View/download PDF
22. Data from Prostate-Specific Antigen Modulates Genes Involved in Bone Remodeling and Induces Osteoblast Differentiation of Human Osteosarcoma Cell Line SaOS-2
- Author
-
Allen C. Gao, Haitao Zhang, Jeffrey M. Conroy, Jason S. Kirk, Farideh Mehraein-Ghomi, Soo Ok Lee, Wei Lou, and Nagalakshmi Nadiminty
- Abstract
Purpose: The high prevalence of osteoblastic bone metastases in prostate cancer involves the production of osteoblast-stimulating factors by prostate cancer cells. Prostate-specific antigen (PSA) is a serine protease uniquely produced by prostate cancer cells and is an important serologic marker for prostate cancer. In this study, we examined the role of PSA in the induction of osteoblast differentiation.Experimental Design: Human cDNA containing a coding region for PSA was transfected into human osteosarcoma SaOS-2 cells. SaOS-2 cells were also treated with exogenously added PSA. We evaluated changes in global gene expression using cDNA arrays and Northern blot analysis resulting from expression of PSA in human osteosarcoma SaOS-2 cells.Results: SaOS-2 cells expressing PSA had markedly up-regulated expression of genes associated with osteoblast differentiation including runx-2 and osteocalcin compared with the controls. Consistent with these results, the stable clones expressing PSA showed increased mineralization and increased activity of alkaline phosphatase in vitro compared with controls, suggesting that these cells undergo osteoblast differentiation. We also found that osteoprotegerin expression was down-regulated and that the receptor activator of NF-κB ligand expression was up-regulated in cells expressing PSA compared with controls.Conclusions: Modulation of the expression of osteogenic genes and alteration of the balance between osteoprotegerin–receptor activator of NF-κB ligand by PSA suggests that PSA produced by metastatic prostate cancer cells may participate in bone remodeling in favor of the development of osteoblastic metastases in the heterogeneous mixture of osteolytic and osteoblastic lesions. These findings provide a molecular basis for understanding the high prevalence of osteoblastic bone metastases in prostate cancer.
- Published
- 2023
- Full Text
- View/download PDF
23. Supplementary tables 1s-4s from The Impact of DNA Input Amount and DNA Source on the Performance of Whole-Exome Sequencing in Cancer Epidemiology
- Author
-
Song Yao, Song Liu, Christine B. Ambrosone, Lawrence H. Kushi, Janise M. Roh, Isaac J. Ergas, Marilyn L. Kwan, Warren Davis, Sean T. Glenn, Jeffrey M. Conroy, Carl D. Morrison, Lei Wei, Jianmin Wang, Lori Shepherd, Qiang Hu, and Qianqian Zhu
- Abstract
Supplementary tables 1s-4s. Supplementary Table 1s. Concordance of variant calls. Supplementary Table 2s. Breast cancer-related genes from the Cancer Gene Consensus database. Supplementary Table 3s. Concordance of variant calls in known breast cancer genes. Supplementary Table 4s. Discordant variant calls in breast cancer-related genes.
- Published
- 2023
- Full Text
- View/download PDF
24. Data from The Impact of DNA Input Amount and DNA Source on the Performance of Whole-Exome Sequencing in Cancer Epidemiology
- Author
-
Song Yao, Song Liu, Christine B. Ambrosone, Lawrence H. Kushi, Janise M. Roh, Isaac J. Ergas, Marilyn L. Kwan, Warren Davis, Sean T. Glenn, Jeffrey M. Conroy, Carl D. Morrison, Lei Wei, Jianmin Wang, Lori Shepherd, Qiang Hu, and Qianqian Zhu
- Abstract
Background: Whole-exome sequencing (WES) has recently emerged as an appealing approach to systematically study coding variants. However, the requirement for a large amount of high-quality DNA poses a barrier that may limit its application in large cancer epidemiologic studies. We evaluated the performance of WES with low input amount and saliva DNA as an alternative source material.Methods: Five breast cancer patients were randomly selected from the Pathways Study. From each patient, four samples, including 3 μg, 1 μg, and 0.2 μg blood DNA and 1 μg saliva DNA, were aliquoted for library preparation using the Agilent SureSelect Kit and sequencing using Illumina HiSeq2500. Quality metrics of sequencing and variant calling, as well as concordance of variant calls from the whole exome and 21 known breast cancer genes, were assessed by input amount and DNA source.Results: There was little difference by input amount or DNA source on the quality of sequencing and variant calling. The concordance rate was about 98% for single-nucleotide variant calls and 83% to 86% for short insertion/deletion calls. For the 21 known breast cancer genes, WES based on low input amount and saliva DNA identified the same set variants in samples from a same patient.Conclusions: Low DNA input amount, as well as saliva DNA, can be used to generate WES data of satisfactory quality.Impact: Our findings support the expansion of WES applications in cancer epidemiologic studies where only low DNA amount or saliva samples are available. Cancer Epidemiol Biomarkers Prev; 24(8); 1207–13. ©2015 AACR.
- Published
- 2023
- Full Text
- View/download PDF
25. Supplementary Table 1 from Prostate-Specific Antigen Modulates Genes Involved in Bone Remodeling and Induces Osteoblast Differentiation of Human Osteosarcoma Cell Line SaOS-2
- Author
-
Allen C. Gao, Haitao Zhang, Jeffrey M. Conroy, Jason S. Kirk, Farideh Mehraein-Ghomi, Soo Ok Lee, Wei Lou, and Nagalakshmi Nadiminty
- Abstract
Supplementary Table 1 from Prostate-Specific Antigen Modulates Genes Involved in Bone Remodeling and Induces Osteoblast Differentiation of Human Osteosarcoma Cell Line SaOS-2
- Published
- 2023
- Full Text
- View/download PDF
26. Supplementary Figure 2s from The Impact of DNA Input Amount and DNA Source on the Performance of Whole-Exome Sequencing in Cancer Epidemiology
- Author
-
Song Yao, Song Liu, Christine B. Ambrosone, Lawrence H. Kushi, Janise M. Roh, Isaac J. Ergas, Marilyn L. Kwan, Warren Davis, Sean T. Glenn, Jeffrey M. Conroy, Carl D. Morrison, Lei Wei, Jianmin Wang, Lori Shepherd, Qiang Hu, and Qianqian Zhu
- Abstract
Supplementary Figure 2s. Comparisons of quality metrics between concordant and discordant short insertion/deletions (indel).
- Published
- 2023
- Full Text
- View/download PDF
27. Supplementary Figure 1s from The Impact of DNA Input Amount and DNA Source on the Performance of Whole-Exome Sequencing in Cancer Epidemiology
- Author
-
Song Yao, Song Liu, Christine B. Ambrosone, Lawrence H. Kushi, Janise M. Roh, Isaac J. Ergas, Marilyn L. Kwan, Warren Davis, Sean T. Glenn, Jeffrey M. Conroy, Carl D. Morrison, Lei Wei, Jianmin Wang, Lori Shepherd, Qiang Hu, and Qianqian Zhu
- Abstract
Supplementary Figure 1s. Comparisons of quality metrics between concordant and discordant single nucleotide variants (SNVs).
- Published
- 2023
- Full Text
- View/download PDF
28. SCNVSim: somatic copy number variation and structure variation simulator.
- Author
-
Maochun Qin, Biao Liu, Jeffrey M. Conroy, Carl D. Morrison, Qiang Hu, Yubo Cheng, Mitsuko Murakami, Adekunle O. Odunsi, Candace S. Johnson, Lei Wei, Song Liu, and Jianmin Wang 0014
- Published
- 2015
- Full Text
- View/download PDF
29. Aligning tumor mutational burden (TMB) quantification across diagnostic platforms: phase II of the Friends of Cancer Research TMB Harmonization Project
- Author
-
Jan Budczies, David Fabrizio, H. Mellert, Mark Li, M. Butler, Phillip Stafford, K. Eyring, Diana Merino Vega, Mark Stewart, Dinesh Cyanam, Kristen Meier, Chen Zhao, Paul M. Williams, Justin Newberg, Warren Tom, Sarabjot Pabla, Ethan Sokol, A. Stenzinger, V.R. Gregersen, Vincent Funari, P. Beer, Roberto Salgado, Lisa M. McShane, G. Pestano, Jen-Hao Cheng, L.K. Bruce, Mingchao Xie, Laura M. Yee, J. Carl Barrett, Jeffrey M. Conroy, Laura Lasiter, R. Samara, Victor J. Weigman, George Green, Jonathan F. Baden, Tomas Vilimas, Jeff Allen, Matthew D. Hellmann, K.C. Valkenburg, Ahmet Zehir, A. J. Lazar, Elizabeth P. Garcia, Laura E. MacConaill, Laurel Keefer, Shu-Jen Chen, X.Z. Wang, Li Chen, A. Pallavajjalla, Yingdong Zhao, and Q. Xie
- Subjects
education.field_of_study ,business.industry ,Software tool ,Population ,Reproducibility of Results ,Hematology ,Computational biology ,Tumor Burden ,Minor allele frequency ,Oncology ,Neoplasms ,Cancer genome ,Mutation ,Atlas data ,Biomarkers, Tumor ,Humans ,Medicine ,education ,business ,Exome - Abstract
Background Tumor mutational burden (TMB) measurements aid in identifying patients who are likely to benefit from immunotherapy; however, there is empirical variability across panel assays and factors contributing to this variability have not been comprehensively investigated. Identifying sources of variability can help facilitate comparability across different panel assays, which may aid in broader adoption of panel assays and development of clinical applications. Materials and methods Twenty-nine tumor samples and 10 human-derived cell lines were processed and distributed to 16 laboratories; each used their own bioinformatics pipelines to calculate TMB and compare to whole exome results. Additionally, theoretical positive percent agreement (PPA) and negative percent agreement (NPA) of TMB were estimated. The impact of filtering pathogenic and germline variants on TMB estimates was assessed. Calibration curves specific to each panel assay were developed to facilitate translation of panel TMB values to whole exome sequencing (WES) TMB values. Results Panel sizes >667 Kb are necessary to maintain adequate PPA and NPA for calling TMB high versus TMB low across the range of cut-offs used in practice. Failure to filter out pathogenic variants when estimating panel TMB resulted in overestimating TMB relative to WES for all assays. Filtering out potential germline variants at >0% population minor allele frequency resulted in the strongest correlation to WES TMB. Application of a calibration approach derived from The Cancer Genome Atlas data, tailored to each panel assay, reduced the spread of panel TMB values around the WES TMB as reflected in lower root mean squared error (RMSE) for 26/29 (90%) of the clinical samples. Conclusions Estimation of TMB varies across different panels, with panel size, gene content, and bioinformatics pipelines contributing to empirical variability. Statistical calibration can achieve more consistent results across panels and allows for comparison of TMB values across various panel assays. To promote reproducibility and comparability across assays, a software tool was developed and made publicly available.
- Published
- 2021
- Full Text
- View/download PDF
30. Visceral Obesity Promotes Lung Cancer Progression—Toward Resolution of the Obesity Paradox in Lung Cancer
- Author
-
Shrunjal Shah, Sai Yendamuri, Charles Roche, Jeffrey M. Conroy, R J Seager, Randall J. Smith, Eric Kannisto, Kris Attwood, Santosh K. Patnaik, Rohit Gosain, Robert Zollo, Stephanie N. Sass, Joseph Barbi, Xialong Wang, Aravind Srinivasan, Cara Petrucci, and Sarabjot Pabla
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,medicine.medical_specialty ,Lung Neoplasms ,Article ,Body Mass Index ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Obesity ,Adverse effect ,Lung cancer ,Retrospective Studies ,business.industry ,Confounding ,Cancer ,medicine.disease ,Metformin ,030104 developmental biology ,Obesity, Abdominal ,030220 oncology & carcinogenesis ,Neoplasm Recurrence, Local ,business ,Body mass index ,Obesity paradox ,medicine.drug - Abstract
Introduction Although obesity is associated with adverse cancer outcomes in general, most retrospective clinical studies suggest a beneficial effect of obesity in NSCLC. Methods Hypothesizing that this “obesity paradox” arises partly from the limitations of using body mass index (BMI) to measure obesity, we quantified adiposity using preoperative computed tomography images. This allowed the specific determination of central obesity as abdominal visceral fat area normalized to total fat area (visceral fat index [VFI]). In addition, owing to the previously reported salutary effect of metformin on high-BMI patients with lung cancer, metformin users were excluded. We then explored associations between visceral obesity and outcomes after surgical resection of stage I and II NSCLC. We also explored potential immunologic underpinnings of such association using complimentary analyses of tumor gene expression data from NSCLC tumors and the tumor transcriptome and immune microenvironment in an immunocompetent model of lung cancer with diet-induced obesity. Results We found that in 513 patients with stage I and II NSCLC undergoing lobectomy, a high VFI is associated with decreased recurrence-free and overall survival. VFI was also inversely related to an inflammatory transcriptomic signature in NSCLC tumors, consistent with observations made in immunocompetent murine models wherein diet-induced obesity promoted cancer progression while exacerbating elements of immune suppression in the tumor niche. Conclusions In all, this study uses multiple lines of evidence to reveal the adverse effects of visceral obesity in patients with NSCLC, which align with those found in animal models. Thus, the obesity paradox may, at least in part, be secondary to the use of BMI as a measure of obesity and the confounding effects of metformin use.
- Published
- 2021
- Full Text
- View/download PDF
31. Integration of tumor inflammation, cell proliferation, and traditional biomarkers improves prediction of immunotherapy resistance and response
- Author
-
Jonathan Andreas, Vincent Giamo, Blake Burgher, Yirong Wang, Margot Schoenborn, Roger Klein, Erik Van Roey, Mary Nesline, Shengle Zhang, Felicia L. Lenzo, Shuang Gao, Sarabjot Pabla, Paul DePietro, R J Seager, Jeffrey M. Conroy, Sean T. Glenn, and Carrie Hoefer
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T cell ,Clinical Biochemistry ,Context (language use) ,RM1-950 ,Algorithmic analysis ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,Borderline ,Medicine ,Inflamed ,Cell proliferation ,Inflammation ,Tumor microenvironment ,business.industry ,Melanoma ,Research ,Biochemistry (medical) ,Immunotherapy ,Gene signature ,medicine.disease ,Ipilimumab ,030104 developmental biology ,medicine.anatomical_structure ,Nivolumab ,030220 oncology & carcinogenesis ,Non-inflamed ,Cancer research ,Molecular Medicine ,Therapeutics. Pharmacology ,business ,Pembrolizumab - Abstract
Background Contemporary to the rapidly evolving landscape of cancer immunotherapy is the equally changing understanding of immune tumor microenvironments (TMEs) which is crucial to the success of these therapies. Their reliance on a robust host immune response necessitates clinical grade measurements of immune TMEs at diagnosis. In this study, we describe a stable tumor immunogenic profile describing immune TMEs in multiple tumor types with ability to predict clinical benefit from immune checkpoint inhibitors (ICIs). Methods A tumor immunogenic signature (TIGS) was derived from targeted RNA-sequencing (RNA-seq) and gene expression analysis of 1323 clinical solid tumor cases spanning 35 histologies using unsupervised analysis. TIGS correlation with ICI response and survival was assessed in a retrospective cohort of NSCLC, melanoma and RCC tumor blocks, alone and combined with TMB, PD-L1 IHC and cell proliferation biomarkers. Results Unsupervised clustering of RNA-seq profiles uncovered a 161 gene signature where T cell and B cell activation, IFNg, chemokine, cytokine and interleukin pathways are over-represented. Mean expression of these genes produced three distinct TIGS score categories: strong (n = 384/1323; 29.02%), moderate (n = 354/1323; 26.76%), and weak (n = 585/1323; 44.22%). Strong TIGS tumors presented an improved ICI response rate of 37% (30/81); with highest response rate advantage occurring in NSCLC (ORR = 36.6%; 16/44; p = 0.051). Similarly, overall survival for strong TIGS tumors trended upward (median = 25 months; p = 0.19). Integrating the TIGS score categories with neoplastic influence quantified via cell proliferation showed highly proliferative and strong TIGS tumors correlate with significantly higher ICI ORR than poorly proliferative and weak TIGS tumors [14.28%; p = 0.0006]. Importantly, we noted that strong TIGS and highly [median = not achieved; p = 0.025] or moderately [median = 16.2 months; p = 0.025] proliferative tumors had significantly better survival compared to weak TIGS, highly proliferative tumors [median = 7.03 months]. Importantly, TIGS discriminates subpopulations of potential ICI responders that were considered negative for response by TMB and PD-L1. Conclusions TIGS is a comprehensive and informative measurement of immune TME that effectively characterizes host immune response to ICIs in multiple tumors. The results indicate that when combined with PD-L1, TMB and cell proliferation, TIGS provides greater context of both immune and neoplastic influences on the TME for implementation into clinical practice.
- Published
- 2021
32. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy
- Author
-
Yu Fujiwara, Shumei Kato, Mary K Nesline, Jeffrey M Conroy, Paul DePietro, Sarabjot Pabla, and Razelle Kurzrock
- Subjects
Class I Phosphatidylinositol 3-Kinases ,Programmed Cell Death 1 Receptor ,Tryptophan ,General Medicine ,B7-H1 Antigen ,Tryptophan Oxygenase ,Oncology ,Receptors, Aryl Hydrocarbon ,Tumor Microenvironment ,Humans ,Indoleamine-Pyrrole 2,3,-Dioxygenase ,Radiology, Nuclear Medicine and imaging ,CTLA-4 Antigen ,Immunotherapy ,Enzyme Inhibitors ,Immune Checkpoint Inhibitors ,Melanoma ,Kynurenine - Abstract
Strategies for unlocking immunosuppression in the tumor microenvironment have been investigated to overcome resistance to first-generation immune checkpoint blockade with anti- programmed cell death protein 1 (PD-1)/ programmed death-ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4) agents. Indoleamine 2,3-dioxygenase (IDO) 1, an enzyme catabolizing tryptophan to kynurenine, creates an immunosuppressive environment in preclinical studies. Early phase clinical trials investigating inhibition of IDO1, especially together with checkpoint blockade, provided promising results. Unfortunately, the phase 3 trial of the IDO1 inhibitor epacadostat combined with the PD-1 inhibitor pembrolizumab did not show clinical benefit when compared with pembrolizumab monotherapy in patients with advanced malignant melanoma, which dampened enthusiasm for IDO inhibitors. Even so, several molecules, such as the aryl hydrocarbon receptor and tryptophan 2,3-dioxygenase, were reported as additional potential targets for the modulation of the tryptophan pathway, which might enhance clinical effectiveness. Furthermore, the combination of IDO pathway blockade with agents inhibiting other signals, such as those generated by PIK3CA mutations that may accompany IDO1 upregulation, may be a novel way to enhance activity. Importantly, IDO1 expression level varies by tumor type and among patients with the same tumor type, suggesting that patient selection based on expression levels of IDO1 may be warranted in clinical trials.
- Published
- 2022
33. Abstract P5-02-54: Application of low-pass whole genome sequencing for the detection of Homologous Recombination Deficiency in breast cancer
- Author
-
Gillian Belbin, Jie An, Chase Mazur, Joseph Pickrell, Jeremy Li, Daniel Metzger, Shuang Gao, Erik Van Roey, Robert Seager, Sarabjot Pabla, Durga Prasad Dash, and Jeffrey M. Conroy
- Subjects
Cancer Research ,Oncology - Abstract
Background: Homologous recombination deficiency (HRD), broadly defined as a loss of the cellular mechanism underlying homologous recombination, is often observed in breast cancer. HRD causes distinctive perturbations to tumor genomic architecture that allow for its molecular identification, while also rendering HRD+ cancers vulnerable to specific chemotherapeutic interventions. This makes the molecular identification of HRD a promising avenue in precision medicine of breast cancer. Specific features of HRD include the presence of large-scale transitions (LST), telomeric allelic imbalance (TAI) and Loss of Heterozygosity (LOH). Each are readily detectable via targeted next generation sequencing (tNGS) or via array-based genotyping. However, genome-wide approaches for HRD detection using cost-effective methods, such as low-pass sequencing (LP-WGS), remain relatively under-explored. Here, we investigated whether HRD signals can be successfully re-capitulated using LP-WGS technology and benchmarked our results against the current field standard (both tNGS and array genotyping). Methods: LP-WGS and tNGS was performed on 96 samples across a range of tumor types (including N=17 breast cancer samples). LP-WGS libraries were prepared using Nextera (Illumina) using 0.4ng DNA input, and sequenced to 0.5-1x coverage. tNGS libraries were prepared using TSO500 (Illumina) using 40-80ng input, and sequenced to >150x unique read coverage. Regions of CNV were estimated using CNVKit v0.9.6, and regions of LOH were estimated using a novel ancestry-aware method. Small variant detection was performed using the TSO500 v2.2.0.12 analysis pipeline. SNP array analysis of 12 tumor samples using Oncoscan (ThermoFisher) was also performed. CNV and LOH estimates derived from LP-WGS, TSO500 and SNP array data were calculated using Jaccard similarity, treating the SNP array data as the “ground truth”. Results: We benchmarked HRD signals derived from LP-WGS compared to the array-based calls and observed near perfect sensitivity for CNV gains across samples (Jaccard index=1.0), as well as for CNV losses between LP-WGS and SNP array (Jaccard index=1.0). We additionally noted that LPS-WGS calls captured both CNV loss and gains that were not detectable via the SNP array. For TAI, LP-WGS re-capitulated 7/10 unique signals also identified via array. We also observed high concordance between regions of the genome called LOH between both platforms (median Jaccard index=0.70, IQR=0.254), but noted an attenuation of sensitivity in samples where estimated tumor heterogeneity was high. We also evaluated LP-WGS CNV calls against the TSO500 assay and noted high sensitivity (96%; 94%) and specificity (89%; 91%) for both CNV gains and losses, respectively. Conclusions: Workflows incorporating LP-WGS can support the detection of HRD genome-wide, paving the way for a more affordable assay that may help to inform clinical decision making in the future treatment of breast cancer. Citation Format: Gillian Belbin, Jie An, Chase Mazur, Joseph Pickrell, Jeremy Li, Daniel Metzger, Shuang Gao, Erik Van Roey, Robert Seager, Sarabjot Pabla, Durga Prasad Dash, Jeffrey M. Conroy. Application of low-pass whole genome sequencing for the detection of Homologous Recombination Deficiency in breast cancer [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P5-02-54.
- Published
- 2023
- Full Text
- View/download PDF
34. Tumor Inflammation, Obesity, and Proliferative Status as Biomarkers in Gastroesophageal Adenocarcinoma
- Author
-
Sarbajit Mukherjee, R. J. Seager, Yong Hee Lee, Jeffrey M. Conroy, Pawel Kalinski, and Sarabjot Pabla
- Subjects
obesity ,biomarker ,gastric cancer ,esophageal cancer ,immunotherapy ,inflammation ,proliferation ,survival ,BMI ,body mass index ,Medicine (miscellaneous) ,Article ,Medicine - Abstract
Recent epidemiological studies have shown that obesity, typically measured by increased body mass index (BMI), is associated with an increased risk of gastroesophageal adenocarcinoma (GEAC), but the contributing molecular and immune mechanisms remain unknown. Since obesity is known to promote chronic inflammation, we hypothesized that obesity leads to inflammation-related immune dysfunction, which can be reversed by immune-modulating therapy. To test our hypothesis, we examined the clinical and molecular data from advanced GEAC patients. To this end, 46 GEAC tumors were evaluated for biomarkers representing tumor inflammation, cell proliferation, and PD-L1 expression. A CoxPH regression model with potential co-variates, followed by pairwise post hoc analysis, revealed that inflammation in the GEAC tumor microenvironment is associated with improved overall survival, regardless of BMI. We also observed a significant association between cell proliferation and progression-free survival in overweight individuals who received immune-modulating therapy. In conclusion, our data confirm the role of the immune system in the natural course of GEAC and its responses to immunotherapies, but do not support the role of BMI as an independent clinically relevant biomarker in this group of patients.
- Published
- 2021
- Full Text
- View/download PDF
35. 77 Prevalence of secondary immunotherapeutic targets in the absence of established immune biomarkers in solid tumors
- Author
-
Vincent Giamo, Blake Burgher, Mary Nesline, Yong Hee Lee, Jeffrey M. Conroy, Roger Klein, Paul DePietro, R J Seager, Sarabjot Pabla, Erik Van Roey, Sean T. Glenn, Shuang Gao, and Shengle Zhang
- Subjects
Pharmacology ,Cancer Research ,Immune system ,Oncology ,business.industry ,Immunology ,Molecular Medicine ,Immunology and Allergy ,Medicine ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,business ,RC254-282 - Abstract
BackgroundImmune checkpoint inhibitor-based therapies have achieved impressive success in the treatment of several cancer types. Predictive immune biomarkers, including PD-L1, MSI and TMB are well established as surrogate markers for immune evasion and tumor-specific neoantigens across many tumors. Positive detection across cancer types varies, but overall ~50% of patients test negative for these primary immune markers.1 In this study, we investigated the prevalence of secondary immune biomarkers outside of PD-L1, TMB and MSI.MethodsComprehensive genomic and immune profiling, including PD-L1 IHC, TMB, MSI and gene expression of 395 immune related genes was performed on 6078 FFPE tumors representing 34 cancer types, predominantly composed of lung cancer (36.7%), colorectal cancer (11.9%) and breast cancer (8.5%). Expression levels by RNA-seq of 36 genes targeted by immunotherapies in solid tumor clinical trials, identified as secondary immune biomarkers, were ranked against a reference population. Genes with a rank value ≥75th percentile were considered high and values were associated with PD-L1 (positive ≥1%), MSI (MSI-H or MSS) and TMB (high ≥10 Mut/Mb) status. Additionally, secondary immune biomarker status was segmented by tumor type and cancer immune cycle roles.ResultsIn total, 41.0% of cases were PD-L1+, 6.4% TMB+, and 0.1% MSI-H. 12.6% of cases were positive for >2 of these markers while 39.9% were triple negative (PD-L1-/TMB-/MSS). Of the PD-L1-/TMB-/MSS cases, 89.1% were high for at least one secondary immune biomarker, with 69.3% having ≥3 markers. PD-L1-/TMB-/MSS tumor types with ≥50% prevalence of high secondary immune biomarkers included brain, prostate, kidney, sarcoma, gallbladder, breast, colorectal, and liver cancer. High expression of cancer testis antigen secondary immune biomarkers (e.g., NY-ESO-1, LAGE-1A, MAGE-A4) was most commonly observed in bladder, ovarian, sarcoma, liver, and prostate cancer (≥15%). Tumors demonstrating T-cell priming (e.g., CD40, OX40, CD137), trafficking (e.g., TGFB1, TLR9, TNF) and/or recognition (e.g., CTLA4, LAG3, TIGIT) secondary immune biomarkers were most represented by kidney, gallbladder, and sarcoma (≥40%), with melanoma, esophageal, head & neck, cervical, stomach, and lung cancer least represented (≥15%).ConclusionsOur studies show comprehensive tumor profiling that includes gene expression can detect secondary immune biomarkers targeted by investigational therapies in ~90% of PD-L1-/TMB-/MSS cases. While genomic profiling could also provide therapeutic choices for a percentage of these patients, detection of secondary immune biomarkers by RNA-seq provides additional options for patients without a clear therapeutic path as determined by PD-L1 testing and genomic profiling alone.ReferenceHuang R S P, Haberberger J, Severson E, et al. A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases. Mod Pathol 2021;34: 252–263.
- Published
- 2021
36. Haplotype Analysis of the T-Cell Receptor Beta (TCRB) Locus by Long-amplicon TCRB Repertoire Sequencing
- Author
-
Jianping Zheng, Linghua Wang, Jeffrey M. Conroy, Denise Topacio-Hall, Dzifa Y. Duose, Carl Morrison, Coya Tapia, Mingxuan Xu, Joud Hajjar, Bettzy Stephen, Lauren Miller, Lifeng Lin, Timothy Looney, Funda Meric-Bernstam, Fiona Hyland, Geoffrey Lowman, Jing Gong, Aung Naing, Alexander Glavin, Elizabeth Linch, and Anas Alshawa
- Subjects
0301 basic medicine ,Whole genome sequencing ,Genetics ,Cancer Research ,Repertoire ,Immunology ,Haplotype ,Locus (genetics) ,Immunogenetics ,Biology ,Amplicon ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Immunology and Allergy ,Allele ,Gene ,030215 immunology - Abstract
Background: Polymorphism within the human T-cell receptor beta variable (TRBV) gene has been proposed as a risk factor for autoimmune disease and immune-related adverse events (IRAEs) during immunotherapy. Previous efforts to evaluate TRBV polymorphism by whole genome sequencing have been hampered by the repetitive nature of the T-cell receptor beta (TCRB) locus. We present a novel long-amplicon TCRB repertoire sequencing approach to enable TRBV haplotype analysis from peripheral blood. Methods: Peripheral blood leukocyte total RNA from 81 Caucasians was used for sequencing of TCRB chains via the Oncomine TCRB-LR assay (amplicon spanning CDR1, 2 and 3) and the Ion Gene Studio S5. VDJ rearrangements were annotated by comparison to the IMGT database, then mined to construct TRBV allele profiles for each individual including, where detected, novel alleles not present in the ImMunoGeneTics (IMGT) database. Finally, TRBV allele profiles were subjected to principal component analysis and k-means clustering to identify TRBV allele haplotypes. Results: Clustering analysis revealed the presence of six major sets of coincident TRBV alleles, which we term haplotype groups. Allelic diversity varied markedly across haplotype groups, with approximately one third of the cohort showing limited TRBV allelic diversity and few uncommon alleles compared to members of other groups. Analysis revealed 37 putatively novel TRBV alleles that are absent from the IMGT database. Conclusion: We demonstrate a straightforward and cost-efficient method for TRBV haplotype analysis from long-amplicon TCRB sequencing data.
- Published
- 2019
- Full Text
- View/download PDF
37. Oncologist uptake of comprehensive genomic profile guided targeted therapy
- Author
-
Jeffrey M. Conroy, Grace K. Dy, Mateusz Opyrchal, Sean T. Glenn, Patrick McKay Boland, Antonios Papanicolau-Sengos, Mark Gardner, Shashikant Lele, S.N. Akers, Carl Morrison, Amy P. Early, Paul DePietro, Peter J. Frederick, Marc S. Ernstoff, Kunle Odunsi, Igor Puzanov, Hongbin Chen, Stephen B. Edge, Mary Nesline, Anne Grand'Maison, Felicia L. Lenzo, and Gurkamal Chatta
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.medical_treatment ,clinical decision making ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,Medical history ,comprehensive genomic profiling ,business.industry ,Cancer ,Immunotherapy ,Guideline ,targeted therapy ,medicine.disease ,real world data ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cohort ,Genomic Profile ,next-generation sequencing ,business ,Research Paper ,Companion diagnostic - Abstract
We describe the extent to which comprehensive genomic profiling (CGP) results were used by oncologists to guide targeted therapy selection in a cohort of solid tumor patients tested as part of standard care at Roswell Park Comprehensive Cancer Center June 2016–June 2017, with adequate follow up through September 2018 (n = 620). Overall, 28.4% of CGP tests advised physicians about targeted therapy use supported by companion diagnostic or practice guideline evidence. Post-test targeted therapy uptake was highest for patients in active treatment at the time of order (86% versus 76% of treatment naïve patients), but also took longer to initiate (median 50 days versus 7 days for treatment naïve patients), with few patients (2.6%) receiving targeted agents prior to testing. 100% of patients with resistance variants did not receive targeted agents. Treatment naïve patients received immunotherapy as the most common alternative. When targeted therapy given off-label or in a trial was the best CGP option, (7%) of patients received it. Our data illustrate the appropriate and heterogeneous use of CGP by oncologists as a longitudinal treatment decision tool based on patient history and treatment needs, and that some patients may benefit from testing prior to initiation of other standard treatments.
- Published
- 2019
- Full Text
- View/download PDF
38. Identification of targets for prostate cancer immunotherapy
- Author
-
Sarabjot Pabla, Yuanquan Yang, Gurkamal Chatta, Sean T. Glenn, Antonios Papanicolau-Sengos, Mary Nesline, Felicia L. Lenzo, Paul DePietro, Shumei Kato, Razelle Kurzrock, Jeffrey M. Conroy, and Carl Morrison
- Subjects
Male ,0301 basic medicine ,Urology ,medicine.medical_treatment ,CD226 ,Nectins ,In situ hybridization ,Biology ,B7-H1 Antigen ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,TIGIT ,Antigens, CD ,Tumor Microenvironment ,medicine ,Humans ,RNA, Neoplasm ,Gene ,Aged ,TOR Serine-Threonine Kinases ,Prostatic Neoplasms ,Microsatellite instability ,Immunotherapy ,Middle Aged ,Programmed Cell Death 1 Ligand 2 Protein ,medicine.disease ,Immunohistochemistry ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Microsatellite Instability - Abstract
Background We performed profiling of the immune microenvironment of castration-resistant (CRPC) and castration-sensitive (CSPC) prostate cancer (PC) in order to identify novel targets for immunotherapy. Methods PD-L1 and CD3/CD8 immunohistochemistry, PD-L1/2 fluorescent in situ hybridization, tumor mutation burden, microsatellite instability, and RNA-seq of 395 immune-related genes were performed in 19 CRPC and CSPC. Targeted genomic sequencing and fusion analysis were performed in 17 of these specimens. Results CD276, PVR, and NECTIN2 were highly expressed in PC. Comparison of CRPC versus CSPC and primary versus metastatic tissue revealed the differential expression of immunostimulatory, immunosuppressive, and epithelial-to-mesenchymal transition (EMT)-related genes. Unsupervised clustering of differentially expressed genes yielded two final clusters best segregated by CRPC and CSPC status. Conclusion CD276 and the alternative checkpoint inhibition PVR/NECTIN2/CD226/TIGIT pathway emerged as relevant to PC checkpoint inhibition target development.
- Published
- 2019
- Full Text
- View/download PDF
39. Immune profiling and immunotherapeutic targets in pancreatic cancer
- Author
-
Razelle Kurzrock, Shumei Kato, Boris W. Kuvshinoff, Jeffrey M. Conroy, Carl Morrison, Blake Burgher, Sarabjot Pabla, Sean T. Glenn, Mary Nesline, Felicia L. Lenzo, and Paul DePietro
- Subjects
0301 basic medicine ,Myeloid ,biology ,business.industry ,Tumor-infiltrating lymphocytes ,medicine.medical_treatment ,Immunosuppression ,General Medicine ,Immunotherapy ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,030220 oncology & carcinogenesis ,Pancreatic cancer ,PD-L1 ,Cancer research ,biology.protein ,Medicine ,Original Article ,business - Abstract
Background Immunotherapeutic approaches for pancreatic ductal adenocarcinoma (PDAC) are less successful as compared to many other tumor types. In this study, comprehensive immune profiling was performed in order to identify novel, potentially actionable targets for immunotherapy. Methods Formalin-fixed paraffin embedded (FFPE) specimens from 68 patients were evaluated for expression of 395 immune-related markers (RNA-seq), mutational burden by complete exon sequencing of 409 genes, PD-L1 expression by immunohistochemistry (IHC), pattern of tumor infiltrating lymphocytes (TILs) infiltration by CD8 IHC, and PD-L1/L2 copy number by fluorescent in situ hybridization (FISH). Results The seven classes of actionable genes capturing myeloid immunosuppression, metabolic immunosuppression, alternative checkpoint blockade, CTLA-4 immune checkpoint, immune infiltrate, and programmed cell death 1 (PD-1) axis immune checkpoint, discerned 5 unique clinically relevant immunosuppression expression profiles (from most to least common): (I) combined myeloid and metabolic immunosuppression [affecting 25 of 68 patients (36.8%)], (II) multiple immunosuppressive mechanisms (29.4%), (III) PD-L1 positive (20.6%), (IV) highly inflamed PD-L1 negative (10.3%); and (V) immune desert (2.9%). The Wilcoxon rank-sum test was used to compare the PDAC cohort with a comparison cohort (n=1,416 patients) for the mean expressions of the 409 genes evaluated. Multiple genes including TIM3, VISTA, CCL2, CCR2, TGFB1, CD73, and CD39 had significantly higher mean expression versus the comparison cohort, while three genes (LAG3, GITR, CD38) had significantly lower mean expression. Conclusions This study demonstrates that a clinically relevant unique profile of immune markers can be identified in PDAC and be used as a roadmap for personalized immunotherapeutic decision-making strategies.
- Published
- 2021
40. A scalable high-throughput targeted next-generation sequencing assay for comprehensive genomic profiling of solid tumors
- Author
-
Shuang Gao, Blake Burgher, Shengle Zhang, Sean T. Glenn, Roger Klein, Vincent Giamo, Sarabjot Pabla, Melissa Mallon, Erik Van Roey, Jonathan Andreas, Yirong Wang, Paul DePietro, R J Seager, Mary Nesline, Felicia L. Lenzo, Yong Hee Lee, and Jeffrey M. Conroy
- Subjects
Genomic profiling ,Molecular biology ,Computer science ,Biochemistry ,Turnaround time ,Workflow ,Sequencing techniques ,Neoplasms ,Basic Cancer Research ,Breast Tumors ,Medicine and Health Sciences ,DNA libraries ,DNA sequencing ,Throughput (business) ,Multidisciplinary ,High-Throughput Nucleotide Sequencing ,RNA sequencing ,Genomics ,Nucleic acids ,Oncology ,Scalability ,Medicine ,Microsatellite Instability ,Transcriptome Analysis ,Research Article ,Next-Generation Sequencing ,DNA Copy Number Variations ,Science ,Computational biology ,Sensitivity and Specificity ,Malignant Tumors ,Cancer Genomics ,Genomic Medicine ,Breast Cancer ,Genetics ,Biomarkers, Tumor ,Humans ,Biology and life sciences ,Sequence Analysis, RNA ,Cancers and Neoplasms ,Computational Biology ,Genetic Variation ,Reproducibility of Results ,DNA ,Genome Analysis ,Research and analysis methods ,Molecular biology techniques ,Mutation - Abstract
Timely and accurate identification of molecular alterations in solid tumors is essential for proper management of patients with advanced cancers. This has created a need for rapid, scalable comprehensive genomic profiling (CGP) systems that detect an increasing number of therapeutically-relevant variant types and molecular signatures. In this study, we assessed the analytical performance of the TruSight Oncology 500 High-Throughput assay for detection of somatic alterations from formalin-fixed paraffin-embedded tissue specimens. In parallel, we developed supporting software and automated sample preparation systems designed to process up to 70 clinical samples in a single NovaSeq 6000TM sequencing run with a turnaround time of 99% overall accuracy and precision. Our results demonstrate that the high-throughput CGP assay is a reliable method for accurate detection of molecular alterations in support of precision therapeutics in oncology. The supporting systems and scalable workflow allow for efficient interpretation and prompt reporting of hundreds of patient cancer genomes per week with excellent analytical performance.
- Published
- 2021
41. Abstract 5137: Cancer testis antigen burden: Pan-cancer distribution and survival implications
- Author
-
R.J. Seager, Erik Van Roey, Shuang Gao, Blake Burgher, Paul DePietro, Mary Nesline, Roger Klein, Shengle Zhang, Jeffrey M. Conroy, and Sarabjot Pabla
- Subjects
Cancer Research ,Oncology - Abstract
Purpose of Study: Cancer testis antigens (CTA) are highly immunogenic genes with the ability to cause cancer-specific immune responses when expressed. Their tumor cell-specific expression makes them a key target of natural T cell response, cancer vaccines, immune checkpoint blockade (ICB), and cell-based immunotherapies in a wide range of tumor types. In this study, we assess the pan-cancer distribution and ICB survival association of CTA burden (CTAB) in real-world solid tumors. Procedure: Three tumor sample cohorts were studied: 1) a pan-cancer discovery cohort to develop a low- and high-CTAB cutoff (n=5450, 39 tumor types), 2) a TCGA cohort (n=19923, 32 tumor types) used to validate the classifier based on CTAB distribution and serve as a non-ICB-treated population, and 3) an ICB-treated retrospective cohort to validate the classification on overall survival (OS) (n=242, 3 tumor types). The expression levels of 17 CTA were measured using targeted RNA-Seq of FFPE tumor samples and then ranked against a pan-cancer reference population. CTAB was calculated for each sample, cohort and tumor type as the sum of the 17 CTA gene expression ranks. The discovery cohort median CTAB of 171 was used to classify all three cohorts into high- and low-CTAB groups. OS analysis was performed on the TCGA and ICB-treated cohorts using a CoxPH regression model to determine the Hazard Ratio (HR). Results: The three cohorts demonstrated overlapping single-peak, left-skewed CTAB distribution curves centered at CTAB values between 170 (discovery cohort) and 256 (retrospective cohort). When grouping by tumor types and ordering by median CTAB, the CTAB distributions for tumor types within all three cohorts were comparable. CoxPH regression analysis revealed an association between the CTAB threshold classifier and OS in both the ICB-treated retrospective and non-ICB TCGA cohorts. However, the direction of this association differed between the two cohorts, with high-CTAB samples having better survival (HR=0.936, p=0.076) in the ICB-treated retrospective cohort and worse survival (HR: 1.007, p=0.084) in the non-ICB-treated cohort. Conclusion: Our studies show that the CTAB distribution was maintained across the discovery and TCGA cohorts and a wide range of tumor types, supporting that the CTAB classifier is valid and histology agnostic. Additionally, when evaluating the ICB and non-ICB-treated cohorts, CTAB demonstrated the ability to predict OS, pointing to the utility of ICB in supporting CTA-specific natural immune response. However, further studies are necessary to verify these mechanisms of response to ICB as well as cancer vaccines and cell-based immunotherapies. Additional validation is needed to establish the predictive utility of CTAB alone and in combination with other immune oncology biomarkers for resistance or response. Citation Format: R.J. Seager, Erik Van Roey, Shuang Gao, Blake Burgher, Paul DePietro, Mary Nesline, Roger Klein, Shengle Zhang, Jeffrey M. Conroy, Sarabjot Pabla. Cancer testis antigen burden: Pan-cancer distribution and survival implications [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5137.
- Published
- 2022
- Full Text
- View/download PDF
42. LAG3 transcriptomic expression correlates with high levels of PD-1, PD-L1, PD-L2, and CTLA-4 checkpoints and with high tumor mutational burden across cancers
- Author
-
Jacob J. Adashek, Shumei Kato, Sarabjot Pabla, Mary Nesline, Jeffrey M. Conroy, Vivek Subbiah, Paul DePietro, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
2561 Background: Lymphocyte Activation Gene 3 (LAG3) or CD223 is an immune checkpoint that can be found on various T cells: CD4+, CD8+, regulatory T cells (Tregs), natural killer T cells, natural killer cells, and plasmacytoid dendritic cells. The expression of LAG3 molecule acts to increase T-cell exhaustion, leading to decreased tumor killing as well as an increase in immune suppressive cytokine release. Many clinical trials of LAG3 inhibitors have had modest effects, but recent data suggests that the LAG3 antibody relatlimab together with nivolumab (anti-PD1) provided greater benefit than nivolumab alone in patients with melanoma. Methods: The RNA expression levels of 397 genes in various types of solid tumors from 514 patients seen at the UCSD Moores Cancer Center were analyzed at a CLIA-licensed laboratory, OmniSeq (https://www.omniseq.com/). Following removal of germline variants, synonymous variants, indels and SNVs with < 5% VAF, TMB is reported as mutations/megabase. Transcript abundance was normalized to internal housekeeping gene profiles and ranked (0-100 percentile) in a standardized manner to a reference population of 735 tumors spanning 35 histologies. Odds ratio for high LAG3 expression was calculated and Bonferroni corrected for multiple genes and cancer histologies with > 40 samples. Results: A total of 116 (22.6%) tumors had high LAG3 (≥75) across 32 different histologies. Cancers with the highest proportion of LAG3 were neuroendocrine (47%), uterine (43%), sarcoma (33%), breast (31%), ovarian (30%), pancreatic (24%), lung (20%), stomach (16%), and colorectal (15%). There was significant association for high LAG3 with high PD-L1 (adj P < 0.001), high PD-1 (adj P < 0.0014), high PD-L2 (adj P < 0.0014), high CTLA-4 (adj P < 0.0014), TMB ≥10 mt/mb (adj P = 0.0504). There was no significant association between histologies colorectal (adj P = 0.1834), breast (adj P = NS), ovarian (adj P = NS), pancreatic (adj P = NS), or gender (adj P = 0.272). Conclusions: High LAG3 was found in almost a quarter of tumor samples and significantly associated with other immune checkpoints with FDA-approved drugs. Ongoing studies combining LAG3 inhibitors and specific immune checkpoint inhibitors may yield more clinical benefit if individualized immunomic transcript interrogation is undertaken, rather than population-based approaches without employment of rationally combined agents matched to each patient’s cancer.
- Published
- 2022
- Full Text
- View/download PDF
43. Comprehensive genomic and immune profiling defines immunotherapy treatment in patients with NSCLC with low PD-L1 IHC
- Author
-
Sarabjot Pabla, Robert J. Seager, Mary Nesline, Paul DePietro, Erik Van Roey, Shuang Gao, Shakti Ramkissoon, Lei Deng, Shengle Zhang, Roger David Klein, and Jeffrey M. Conroy
- Subjects
Cancer Research ,Oncology - Abstract
2623 Background: Immune checkpoint inhibitors (ICIs) have emerged as effective treatments in non-small cell lung cancer (NSCLC). While the clinical utility of single agent ICI or in combination with chemotherapy has been well established, there remains an unmet need for the development of biomarkers that can better predict response. To address this need, we developed and applied a combination genomic and immune biomarker strategy to ICI-treated NSCLC patients which identified distinct patient subgroups with differential benefit among single agent or combination ICI treatment strategies. Methods: A discovery cohort (DC) of 5450 tumors across 37 histologies were evaluated by comprehensive genomic and immune profiling of the tumor immune microenvironment. Individual and combination biomarker assessment included PD-L1 IHC, TMB, tumor inflammation (TIGS), cell proliferation (CP) and cancer testis antigen burden (CTAB). From this cohort, combinations of molecular and immune biomarkers were identified and applied to a retrospective cohort (RC) of 225 metastatic NSCLC patients treated with pembrolizumab + chemo or pembrolizumab alone to correlate with response. Comparison of objective response rates (ORR) was performed using Chi-square test. Kaplan-Meir analysis was performed to test for differences in overall survival (OS) and 1-year OS. Results: Unsupervised analysis of the DC revealed four distinct biomarker combination groups that describe underlying tumor immunobiology: tumor dominant (CTAB, TMB, CP High), proliferative (CP High), inflamed (TIGS High), and checkpoint (PDL1, TIGS and TMB High). Application of these biomarker groups to the RC demonstrated significant differences in response to ICI regimens between groups (p = 0.04). Patients in the proliferative group (35.1%, 79/225; median PD-L1 = 20% TPS) treated with single agent pembrolizumab showed a significantly higher ORR (59%; 16/27) compared to pembrolizumab + chemo (27%; 14/52; p = 0.005), significantly improved 1-yr OS (p = 0.03), and trend towards better OS (p = 0.14). Importantly, patients in the inflamed group (16%, 36/225; median PD-L1 = 1% TPS), suggested that pembrolizumab + chemo (ORR 26.1%; 6/23) was not associated with ORR compared to pembrolizumab (ORR 31%; 4/13, p = 0.76), or OS (p = 0.37) and 1-yr OS (p = 0.57). Conclusions: Comprehensive genomic and immune profiling may identify PD-L1 low NSCLC patients who benefit from single agent pembrolizumab. PD-L1 low NSCLC patients with a proliferative phenotype may benefit from single agent pembrolizumab, whereas PD-L1 low cases with an inflamed phenotype may benefit from both single agent and combination pembrolizumab. Although further clinical validation of these predictive biomarker combinations is required, this data-driven approach demonstrates the potential to provide treatment decision support when selecting an ICI therapeutic strategy in lung cancer.
- Published
- 2022
- Full Text
- View/download PDF
44. Comprehensive genomic and immune profiling (CGIP) treatment patterns and survival in non-small cell lung cancer (NSCLC)
- Author
-
Mary Nesline, Sarabjot Pabla, Yong Hee Lee, Paul DePietro, Shengle Zhang, Roger David Klein, Jeffrey M. Conroy, Shakti Ramkissoon, Amy P. Early, Lei Deng, and Grace K. Dy
- Subjects
Cancer Research ,Oncology - Abstract
e21167 Background: CGIP analyzes FFPE tumor tissue by DNA/RNA sequencing for SNVs, indels, copy gain/loss, fusions, splice variants, MSI and TMB, along with PD-L1 IHC. A purported advantage of CGIP in NSCLC is the ability to identify targeted and immunotherapy biomarkers to inform clinical management. However, the extent to which CGIP supports treatment decisions and benefits NSCLC patients in various treatment settings is limited. Methods: A retrospective analysis of OmniSeq CGIP results (June 2017-March 2019) and real-world clinical data (through March 2020) for NSCLC patients (n = 300) was performed to evaluate treatment strategies at Roswell Park Comprehensive Cancer Center. Patient targeted and immunotherapies following CGIP were classified as “matched” to biomarker results (established or potentially clinically significant) at the indication level (single or multi-marker results, histology, treatment line) based on AMP/ASCO/CAP guidance for strength of biomarker clinical evidence. We estimated overall survival (OS) from CGIP report date for patients who first received either matched therapy or chemotherapy (and no subsequent matched therapy), and assessed the predictive value of matched therapy for OS in the first or subsequent line setting, adjusting for clinicopathologic covariates. Results: Most CGIP tested patients were female (55%), stage IIIB/IV (89%), ECOG < 2 (83%), non-squamous (86%), treatment naïve (62%), ever smokers (88%). 74% (228) of patients were treated post-CGIP, with 71% receiving at least one matched therapy. Matched therapies received in the frontline setting were supported by the highest (Tier 1A) category of evidence more often than subsequent line therapies (97% vs. 68%). 90% of patients with oncogenic driver mutations received targeted agents (17% of total) and 57% received matched immunotherapy. In the frontline setting, compared to chemotherapy, OS was highest for patients who first received matched targeted therapy (median = 23.4 mo; HR 0.26; p = .004; 95% CI 0.13-0.68) vs matched immunotherapy (median = 17.9 mo; HR 0.38; p = .001; 95% CI 0.21-0.69). Subsequent line, OS was also highest for patients who first received matched targeted therapy (median not est., mean = 27.5 mo; HR 0.20; p = .063; 95% CI 0.04-1.09) vs matched immunotherapy (median = 17.4 mo; HR 0.20; 95% CI 0.04-1.09), however, these differences were non-significant. Conclusions: CGIP supports evidence-based clinical decision making for NSCLC in the first and subsequent line settings and leads to improved survival for patients who receive matched targeted or immunotherapy compared to chemotherapy. Better predictive markers are needed to identify NSCLC patients who are more likely to respond to immunotherapies. Heterogeneity of patient biomarker profiles and treatment strategies over time in real world practice are a challenge to assessing CGIP efficacy.
- Published
- 2022
- Full Text
- View/download PDF
45. Comprehensive transcriptomic analysis of immune checkpoint markers in a pancancer cohort: Implications for response and resistance
- Author
-
Hirotaka Miyashita, Nicholas J. Bevins, Kartheeswaran Thangathurai, Suzanna Lee, Sarabjot Pabla, Mary Nesline, Sean Glenn, Jeffrey M. Conroy, Paul DePietro, Eitan Rubin, Jason K. Sicklick, Shumei Kato, and Razelle Kurzrock
- Subjects
Cancer Research ,Oncology - Abstract
2555 Background: Although immune checkpoint blockade (ICB) has revolutionized cancer treatment, not all patients with cancer benefit from ICB. One possible explanation for poor responders/resistance is the variable expression level of the target molecules (e.g., PD-1 and PD-L1) in the tumor microenvironment. There are recent or ongoing trials targeting variable pathways for immune evasion (e.g., LAG3 or IDO1). It is therefore of interest to know the expression levels related to variable immune checkpoints so that clinical trials can focus on the patients who can benefit from the cognate treatment. Methods: Overall, 514 patients with various solid tumors seen at the University of San Diego, Moores Center for Personalized Cancer Therapy were analyzed. The expression levels of checkpoint markers (ADORA2A, BTLA, CD276, CTLA4, IDO1, IDO2, LAG3, NOS2, PD-1, PD-L1, PD-L2, PVR, TIGIT, TIM3, VISTA, and VTCN) in the tumor samples were measured through RNA sequencing and normalized to internal housekeeping gene profiles, and ranked from 0 to 100 percentile based on a reference population. The expressions of each checkpoint marker were correlated with cancer types, microsatellite instability (MSI), tumor mutational burden (TMB), and programmed death-ligand 1 (PD-L1) status on immunohistochemistry. Results: In this cohort, 60% were female, median age of 60, and included 30 different tumor types, with colorectal cancer being the most common (27%). The rank values of all checkpoint markers were distributed broadly from 0 to 99 or 100. CD276 and NOS2 had the highest (68th percentile) and lowest (13.5 percentile) median rank values, respectively. When rank values were categorized to “Low” (0-24), “Intermediate” (25-74), and “High” (75-100), 41.6% of patients showed high expression of CD276 while only 13% showed high expression of PD-L1. Each patient had a distinctive protfolio of the categorical expression levels of 16 checkpoint markers. Several checkpoint markers, especially NOS2, showed a significant correlation with cancer type. (median rank values in colorectal, stomach, pancreatic, and breast cancer were 79, 76, 5 and 0 respectively, p < 0.001) Five markers (IDO1, LAG3, PD-1, PD-L1, and TIGIT) showed significant correlation with MSI, while seven markers (CTLA4, IDO1, LAG3, PD-1, PD-L1, PD-L2, and TIGIT) were significantly associated with positive PD-L1 status. However, no significant association was seen based on TMB or tissue-specific grouping of patients. Conclusions: The expression of immune checkpoint markers varies from patient to patient, though transcript expression of several markers correlates with cancer type, MSI, and PD-L1 status. Clinical trials with patient selection based on the expression level of checkpoint markers matched to the corresponding ICB drug are warranted.
- Published
- 2022
- Full Text
- View/download PDF
46. 65 PD-L1 by RNA next generation sequencing: comparison with PD-L1 IHC 22C3 and association with survival benefit from pembrolizumab with or without chemotherapy in non-small cell lung cancer
- Author
-
Paul DePietro, Jeffrey M. Conroy, Mary Nesline, Grace K. Dy, Sarabjot Pabla, and Yong Hee Lee
- Subjects
Oncology ,medicine.medical_specialty ,Chemotherapy ,biology ,business.industry ,medicine.medical_treatment ,Pembrolizumab ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,lcsh:RC254-282 ,Percentile rank ,Internal medicine ,PD-L1 ,medicine ,biology.protein ,Immunohistochemistry ,Liquid biopsy ,business ,Lung cancer ,Companion diagnostic - Abstract
Background PD-L1 immunohistochemistry (IHC) testing is suboptimal for predicting patient clinical benefit for checkpoint inhibition, while PD-L1 liquid biopsy is not clinically validated and lacks sensitivity, underscoring the need to include PD-L1 testing in more robust, tissue-efficient, comprehensive, scalable next generation sequencing (NGS) tests. Methods To assess comparability and efficacy of PD-L1 testing by NGS with IHC, we identified NSCLC patients treated by first-line pembrolizumab alone (n=54) or pembrolizumab + chemotherapy (n=49) whose tumors underwent companion diagnostic PD-L1 testing by IHC antibody 22C3 testing (high≥50%; low=1–49%, or negative=0% tissue proportion score), and also by RNA-seq, as part of a comprehensive immune profiling panel. PD-L1 expression by RNA-seq, was measured as a percentile rank, with ≥75 considered ‘high’, and Results More than 75% of IHC high cases were classified as high by RNA-Seq for both treatment groups (p Conclusions PD-L1 status by RNA-seq and IHC appear to be comparable. Unlike PD-L1 IHC however, PD-L1 RNA-seq high status versus not high status is associated with greater survival benefit, indicating PD-L1 by NGS may have utility for pembrolizumab selection.
- Published
- 2020
- Full Text
- View/download PDF
47. Proliferative potential and response to nivolumab in clear cell renal cell carcinoma patients
- Author
-
Sean T. Glenn, Antonios Papanicolau-Sengos, Jeffrey M. Conroy, Blake Burgher, Keisuke Shirai, Carl Morrison, Rajan T. Gupta, Jason Zhu, Konstantin H. Dragnev, Pooja Ghatalia, Razelle Kurzrock, Mary Nesline, Saby George, Vincent Giamo, Wiam Bshara, Tian Zhang, Felicia L. Lenzo, Katherine G. Madden, Matthew Labriola, Bo Xu, Michelle S Park, Matthew Zibelman, Jonathan Andreas, Rajbir Singh, Robert Edwards, Daniel J. George, Shannon J. McCall, Robin Jacob, Yirong Wang, Laura J. Tafe, Farshid Dayyani, and Sarabjot Pabla
- Subjects
0301 basic medicine ,Male ,Kidney Disease ,Immune checkpoint inhibitors ,medicine.medical_treatment ,urologic and male genital diseases ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Renal cell carcinoma ,PD-1 ,Immunology and Allergy ,Medicine ,RC254-282 ,Original Research ,Cancer ,biology ,ki-67 ,pd-1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Middle Aged ,Progression-Free Survival ,Immunological ,Nivolumab ,Oncology ,030220 oncology & carcinogenesis ,Ki-67 ,Immunohistochemistry ,Female ,Immunotherapy ,Research Article ,Adult ,PD-L1 ,renal cell carcinoma ,proliferation ,Immunology ,Oncology and Carcinogenesis ,Antineoplastic Agents ,03 medical and health sciences ,pd-l1 ,Genetics ,Humans ,Carcinoma, Renal Cell ,Aged ,nivolumab ,business.industry ,Carcinoma ,Renal Cell ,RC581-607 ,medicine.disease ,Clear cell renal cell carcinoma ,030104 developmental biology ,Good Health and Well Being ,Cancer research ,biology.protein ,Immunologic diseases. Allergy ,business - Abstract
Background Biomarkers predicting immunotherapy response in metastatic renal cell cancer (mRCC) are lacking. PD-L1 immunohistochemistry is a complementary diagnostic for immune checkpoint inhibitors (ICIs) in mRCC, but has shown minimal clinical utility and is not used in routine clinical practice. Methods Tumor specimens from 56 patients with mRCC who received nivolumab were evaluated for PD-L1, cell proliferation (targeted RNA-seq), and outcome. Results For 56 patients treated with nivolumab as a standard of care, there were 2 complete responses and 8 partial responses for a response rate of 17.9%. Dividing cell proliferation into tertiles, derived from the mean expression of 10 proliferation-associated genes in a reference set of tumors, poorly proliferative tumors (62.5%) were more common than moderately (30.4%) or highly proliferative (8.9%) counterparts. Moderately proliferative tumors were enriched for PD-L1 positive (41.2%), compared to poorly proliferative counterparts (11.4%). Objective response for moderately proliferative (29.4%) tumors was higher than that of poorly (11.4%) proliferative counterparts, but not statistically significant (p = .11). When cell proliferation and negative PD-L1 tumor proportion scores were combined statistically significant results were achieved (p = .048), showing that patients with poorly proliferative and PD-L1 negative tumors have a very low response rate (6.5%) compared to moderately proliferative PD-L1 negative tumors (30%). Conclusions Cell proliferation has value in predicting response to nivolumab in clear cell mRCC patients, especially when combined with PD-L1 expression. Further studies which include the addition of progression-free survival (PFS) along with sufficiently powered subgroups are required to further support these findings.
- Published
- 2020
48. Analysis of mutations in primary and metastatic synovial sarcoma
- Author
-
Tao Yu, Shinya Tanaka, Carl Morrison, Annie Pao, Akira Kawai, Lei Wei, Y. Eugene Yu, Xiaoling Jiang, Christopher Choi, Wiam Bshara, Sean T. Glenn, Jianmin Wang, Song Liu, Jeffrey M. Conroy, and Zhuo Xing
- Subjects
0301 basic medicine ,synovial sarcoma ,whole exome sequencing ,Metastasis ,Fusion gene ,03 medical and health sciences ,0302 clinical medicine ,Primary Synovial Sarcoma ,medicine ,metastasis ,Missense mutation ,SS18-SSX ,Exome sequencing ,Metastatic Synovial Sarcoma ,business.industry ,Soft tissue sarcoma ,ADAM 17 ,medicine.disease ,Synovial sarcoma ,3. Good health ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,business ,Research Paper - Abstract
Synovial sarcoma is the most common pediatric non-rhabdomyosarcoma soft tissue sarcoma and accounts for about 8–10% of all soft tissue sarcoma in childhood and adolescence. The presence of a chromosomal translocation-associated SS18-SSX-fusion gene is causally linked to development of primary synovial sarcoma. Metastases occur in approximately 50–70% of synovial sarcoma cases with yet unknown mechanisms, which led to about 70–80% mortality rate in five years. To explore the possibilities to investigate metastatic mechanisms of synovial sarcoma, we carried out the first genome-wide search for potential genetic biomarkers and drivers associated with metastasis by comparative mutational profiling of 18 synovial sarcoma samples isolated from four patients carrying the primary tumors and another four patients carrying the metastatic tumors through whole exome sequencing. Selected from the candidates yielded from this effort, we examined the effect of the multiple missense mutations of ADAM17, which were identified solely in metastatic synovial sarcoma. The mutant alleles as well as the wild-type control were expressed in the mammalian cells harboring the SS18-SSX1 fusion gene. The ADAM17-P729H mutation was shown to enhance cell migration, a phenotype associated with metastasis. Therefore, like ADAM17-P729H, other mutations we identified solely in metastatic synovial sarcoma may also have the potential to serve as an entry point for unraveling the metastatic mechanisms of synovial sarcoma.
- Published
- 2018
- Full Text
- View/download PDF
49. OncoKids
- Author
-
Moiz Bootwalla, Joshua L. Deignan, Tracy M. Busse, Gordana Raca, James Done, Jonathan D. Buckley, Chao Jie Zhen, Jaclyn A. Biegel, Xinjie Xu, Xiaowu Gai, Beverly Barton Rogers, David M. Parham, Alexander R. Judkins, Alex Ryutov, Matthew C. Hiemenz, Amanda L. Treece, Dennis T. Maglinte, Timothy J. Triche, Jianling Ji, Jeffrey M. Conroy, Dejerianne Ostrow, and Florette K. Hazard
- Subjects
0301 basic medicine ,RNA ,Biology ,DNA sequencing ,Pathology and Forensic Medicine ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Gene duplication ,Cancer research ,medicine ,Molecular Medicine ,Coding region ,Oncogene Fusion ,Bone marrow ,Gene ,DNA - Abstract
The OncoKids panel is an amplification-based next-generation sequencing assay designed to detect diagnostic, prognostic, and therapeutic markers across the spectrum of pediatric malignancies, including leukemias, sarcomas, brain tumors, and embryonal tumors. This panel uses low input amounts of DNA (20 ng) and RNA (20 ng) and is compatible with formalin-fixed, paraffin-embedded and frozen tissue, bone marrow, and peripheral blood. The DNA content of this panel covers the full coding regions of 44 cancer predisposition loci, tumor suppressor genes, and oncogenes; hotspots for mutations in 82 genes; and amplification events in 24 genes. The RNA content includes 1421 targeted gene fusions. We describe the validation of this panel by using a large cohort of 192 unique clinical samples that included a wide range of tumor types and alterations. Robust performance was observed for analytical sensitivity, reproducibility, and limit of detection studies. The results from this study support the use of OncoKids for routine clinical testing of a wide variety of pediatric malignancies.
- Published
- 2018
- Full Text
- View/download PDF
50. Analytical Validation of a Next-Generation Sequencing Assay to Monitor Immune Responses in Solid Tumors
- Author
-
Angela Omilian, Mark Gardner, Fiona Hyland, Wiam Bshara, Jeffrey M. Conroy, Antonios Papanicolau-Sengos, Sean T. Glenn, Vincent Giamo, Blake Burgher, Ji He, Moachun Qin, Mary Nesline, Carl Morrison, Igor Puzanov, Felicia L. Lenzo, Lorenzo Galluzzi, Jonathan Andreas, Sarabjot Pabla, and Marc S. Ernstoff
- Subjects
0301 basic medicine ,RNA Stability ,Lymphocyte ,Biology ,DNA sequencing ,Pathology and Forensic Medicine ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Limit of Detection ,Neoplasms ,medicine ,Humans ,Tumor microenvironment ,Sequence Analysis, RNA ,Detection threshold ,High-Throughput Nucleotide Sequencing ,Reproducibility of Results ,RNA ,DNA, Neoplasm ,Sequence Analysis, DNA ,Molecular biology ,Gene Expression Regulation, Neoplastic ,genomic DNA ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Molecular Medicine - Abstract
We have developed a next-generation sequencing assay to quantify biomarkers of the host immune response in formalin-fixed, paraffin-embedded (FFPE) tumor specimens. This assay aims to provide clinicians with a comprehensive characterization of the immunologic tumor microenvironment as a guide for therapeutic decisions on patients with solid tumors. The assay relies on RNA-sequencing (seq) to semiquantitatively measure the levels of 43 transcripts related to anticancer immune responses and 11 transcripts that reflect the relative abundance of tumor-infiltrating lymphocytes, as well as on DNA-seq to estimate mutational burden. The assay has a clinically relevant 5-day turnaround time and can be conducted on as little as 2.5 ng of RNA and 1.8 ng of genomic DNA extracted from three to five standard FFPE sections. The standardized next-generation sequencing workflow produced sequencing reads adequate for clinical testing of matched RNA and DNA from several samples in a single run. Assay performance for gene-specific sensitivity, linearity, dynamic range, and detection threshold was estimated across a wide range of actual and artificial FFPE samples selected or generated to address preanalytical variability linked to specimen features (eg, tumor-infiltrating lymphocyte abundance, percentage of necrosis), and analytical variability linked to assay features (eg, batch size, run, day, operator). Analytical precision studies demonstrated that the assay is highly reproducible and accurate compared with established orthogonal approaches.
- 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.