68 results on '"Tricia R Cottrell"'
Search Results
2. Neoadjuvant nivolumab plus ipilimumab in resectable non-small cell lung cancer
- Author
-
David Jones, Taha Merghoub, Hok Yee Chan, Franco Verde, Daphne Wang, James R White, Tricia R Cottrell, Joshua E Reuss, Stephen Broderick, Kellie N Smith, Marianna Zahurak, Mara Lanis, Joseph C Murray, Caroline McCarthy, Stephen Yang, Richard Battafarano, Errol Bush, Malcolm Brock, Jinny Ha, Janis Taube, Peter Illei, Ben Levy, and Jamie E Chaft
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background We conducted the first trial of neoadjuvant PD-1 blockade in resectable non-small cell lung cancer (NSCLC), finding nivolumab monotherapy to be safe and feasible with an encouraging rate of pathologic response. Building on these results, and promising data for nivolumab plus ipilimumab (anti-CTLA-4) in advanced NSCLC, we expanded our study to include an arm investigating neoadjuvant nivolumab plus ipilimumab.Methods Patients with resectable stage IB (≥4 cm)–IIIA (American Joint Committee on Cancer Tumor Node Metastases seventh edition), histologically confirmed, treatment-naïve NSCLC received nivolumab 3 mg/kg intravenously plus ipilimumab 1 mg/kg intravenously 6 weeks prior to planned resection. Nivolumab 3 mg/kg was given again approximately 4 and 2 weeks preoperatively. Primary endpoints were safety and feasibility with a planned enrollment of 15 patients. Pathologic response was a key secondary endpoint.Results While the treatment regimen was feasible per protocol, due to toxicity, the study arm was terminated early by investigator consensus after 9 of 15 patients were enrolled. All patients received every scheduled dose of therapy and were fit for planned surgery; however, 6 of 9 (67%) experienced treatment-related adverse events (TRAEs) and 3 (33%) experienced grade ≥3 TRAEs. Three of 9 patients (33%) had biopsy-confirmed tumor progression precluding definitive surgery. Of the 6 patients who underwent resection, 3 are alive and disease-free, 2 experienced recurrence and are actively receiving systemic treatment, and one died postoperatively due to acute respiratory distress syndrome. Two patients who underwent resection had tumor pathologic complete responses (pCRs) and continue to remain disease-free over 24 months since surgery. Pathologic response correlated with pre-treatment tumor PD-L1 expression, but not tumor mutation burden. Tumor KRAS/STK11 co-mutations were identified in 5 of 9 patients (59%), of whom two with disease progression precluding surgery had tumor KRAS/STK11/KEAP1 co-mutations.Conclusions Though treatment was feasible, due to toxicity the study arm was terminated early by investigator consensus. In light of this, and while the long-term disease-free status of patients who achieved pCR is encouraging, further investigation of neoadjuvant nivolumab plus ipilimumab in patients with resectable NSCLC requires the identification of predictive biomarkers that enrich for response.
- Published
- 2020
- Full Text
- View/download PDF
3. Chronic immune checkpoint inhibitor pneumonitis
- Author
-
Joanne Riemer, Daphne Wang, Tricia R Cottrell, Lonny B Yarmus, K Ranh Voong, Hans Lee, and Franco R D'Alessio
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background Pneumonitis from immune checkpoint inhibitors (ICI) is a potentially fatal immune-related adverse event (irAE) from antiprogrammed death 1/programmed death ligand 1 immunotherapy. Most cases of ICI pneumonitis improve or resolve with 4–6 weeks of corticosteroid therapy. Herein, we report the incidence, clinicopathological features and management of patients with non-small cell lung cancer (NSCLC) and melanoma who developed chronic ICI pneumonitis that warrants ≥12 weeks of immunosuppression.Methods Patients with ICI pneumonitis were identified from institutional databases of ICI-treated patients with advanced melanoma and NSCLC between January 2011 and July 2018. ICI pneumonitis was defined as clinical/radiographic evidence of lung inflammation without alternative diagnoses, adjudicated by a multidisciplinary team. Chronic ICI pneumonitis was defined as pneumonitis that persists or worsens with steroid tapering, and necessitates ≥12 weeks of immunosuppression, after ICI discontinuation. Serial chest CT was used to assess radiological features, and tumor response by Response EvaluationCriteria for Solid Tumors V.1.1. Bronchoalveolar lavage fluid (BALF) samples were assessed by cell differential. Lung biopsy samples were evaluated by H&E staining and multiplex immunofluorescence (mIF), where available.Results Among 299 patients, 44 developed ICI pneumonitis (NSCLC: 5/205; melanoma: 1/94), and of these, 6 experienced chronic ICI pneumonitis. The overall incidence of chronic ICI pneumonitis was thus 2%. Of those who developed chronic ICI pneumonitis: the majority had NSCLC (5/6), all sustained disease control from ICIs, and none had other concurrent irAEs. Timing of chronic ICI pneumonitis development was variable (range: 0–50 months), and occurred at a median of 12 months post ICI start. Recrudescence of ICI pneumonitis occurred at a median of 6 weeks after initial steroid start (range: 3–12 weeks), with all patients requiring steroid reintroduction when tapered to ≤10 mg prednisone/equivalent. The median total duration of steroids was 37 weeks (range: 16–43+weeks). Re-emergence of radiographic ICI pneumonitis occurred in the same locations on chest CT, in most cases (5/6). All patients who developed chronic ICI pneumonitis had BALF lymphocytosis on cell differential and organising pneumonia on lung biopsy at initial ICI pneumonitis presentation, with persistent BALF lymphocytosis and brisk CD8+ infiltration on mIF at pneumonitis re-emergence during steroid taper.Conclusions A subset of patients who develop pneumonitis from ICIs will develop chronic ICI pneumonitis, that warrants long-term immunosuppression of ≥12 weeks, and has distinct clinicopathological features.
- Published
- 2020
- Full Text
- View/download PDF
4. Expression of the Autoantigen Topoisomerase‐1 is Enriched in the Lung Tissues of Patients With Autoimmune Interstitial Lung Disease: A Case Control Study
- Author
-
Tricia R. Cottrell, Frederic Askin, Marc K. Halushka, Livia Casciola‐Rosen, and Zsuzsanna H. McMahan
- Subjects
Diseases of the musculoskeletal system ,RC925-935 - Abstract
Background Among the autoimmune rheumatic diseases, it is striking that autoantibodies targeting ubiquitously expressed proteins (eg, topoisomerase‐1) associate with specific clinical complications (eg, interstitial lung disease [ILD]). It has been proposed that enriched antigen expression in inflamed target tissue may play a role in focusing the autoimmune response. We sought to determine whether topoisomerase‐1 expression is enriched in lungs from patients with autoimmune/inflammatory diseases relative to normal lung. Methods We used a 99‐core lung tissue microarray (TMA) containing lung tissue from 40 patients with autoimmune inflammatory ILD (cases) and 46 control subjects with normal lungs. We stained the TMA with antibodies to compare topoisomerase‐1 and CD8 expression between patients and control subjects and evaluated whether expression is enriched in specific cell types. Staining was analyzed, and statistical comparisons were performed. Results Cases were more likely to have global topoisomerase‐1 expression (53% vs 21%; P = 0.003), specifically in pneumocytes (47% vs 16%; P = 0.003) and stromal/immune cells (32% vs 5%; P = 0.002) compared with control subjects. CD8 cell density (223 cells/mm2 vs 102 cells/mm2; P = 0.018) was significantly higher in topoisomerase‐1–positive lung tissues compared with topoisomerase‐1–negative lung tissues. Interestingly, topoisomerase‐1 expression was significantly more common in scleroderma compared with normal lung (67% vs 21%; P = 0.036) and was present more frequently in pneumocytes in these patients (67% vs 16%; P = 0.018). Conclusions Pulmonary expression of topoisomerase‐1 is increased in the setting of autoimmune ILD relative to normal lung, specifically in pneumocytes. This may contribute to the amplification of pulmonary disease in patients with scleroderma with a loss of tolerance to topoisomerase‐1.
- Published
- 2020
- Full Text
- View/download PDF
5. Persistent mutant oncogene specific T cells in two patients benefitting from anti-PD-1
- Author
-
Kellie N. Smith, Nicolas J. Llosa, Tricia R. Cottrell, Nicholas Siegel, Hongni Fan, Prerna Suri, Hok Yee Chan, Haidan Guo, Teniola Oke, Anas H. Awan, Franco Verde, Ludmila Danilova, Valsamo Anagnostou, Ada J. Tam, Brandon S. Luber, Bjarne R. Bartlett, Laveet K. Aulakh, John-William Sidhom, Qingfeng Zhu, Cynthia L. Sears, Leslie Cope, William H. Sharfman, Elizabeth D. Thompson, Joanne Riemer, Kristen A. Marrone, Jarushka Naidoo, Victor E. Velculescu, Patrick M. Forde, Bert Vogelstein, Kenneth W. Kinzler, Nickolas Papadopoulos, Jennifer N. Durham, Hao Wang, Dung T. Le, Sune Justesen, Janis M. Taube, Luis A. Diaz, Julie R. Brahmer, Drew M. Pardoll, Robert A. Anders, and Franck Housseau
- Subjects
Checkpoint blockade ,Predictive biomarkers ,Oncogene ,Neoantigens ,T cells ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Several predictive biomarkers are currently approved or are under investigation for the selection of patients for checkpoint blockade. Tumor PD-L1 expression is used for stratification of non-small cell lung (NSCLC) patients, with tumor mutational burden (TMB) also being explored with promising results, and mismatch-repair deficiency is approved for tumor site-agnostic disease. While tumors with high PD-L1 expression, high TMB, or mismatch repair deficiency respond well to checkpoint blockade, tumors with lower PD-L1 expression, lower mutational burdens, or mismatch repair proficiency respond much less frequently. Case presentation We studied two patients with unexpected responses to checkpoint blockade monotherapy: a patient with PD-L1-negative and low mutational burden NSCLC and one with mismatch repair proficient colorectal cancer (CRC), both of whom lack the biomarkers associated with response to checkpoint blockade, yet achieved durable clinical benefit. Both maintained T-cell responses in peripheral blood to oncogenic driver mutations – BRAF-N581I in the NSCLC and AKT1-E17K in the CRC – years after treatment initiation. Mutation-specific T cells were also found in the primary tumor and underwent dynamic perturbations in the periphery upon treatment. Conclusions These findings suggest that T cell responses to oncogenic driver mutations may be more prevalent than previously appreciated and could be harnessed in immunotherapeutic treatment, particularly for patients who lack the traditional biomarkers associated with response. Comprehensive studies are warranted to further delineate additional predictive biomarkers and populations of patients who may benefit from checkpoint blockade.
- Published
- 2019
- Full Text
- View/download PDF
6. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab
- Author
-
Nicolas A. Giraldo, Peter Nguyen, Elizabeth L. Engle, Genevieve J. Kaunitz, Tricia R. Cottrell, Sneha Berry, Benjamin Green, Abha Soni, Jonathan D. Cuda, Julie E. Stein, Joel C. Sunshine, Farah Succaria, Haiying Xu, Aleksandra Ogurtsova, Ludmila Danilova, Candice D. Church, Natalie J. Miller, Steve Fling, Lisa Lundgren, Nirasha Ramchurren, Jennifer H. Yearley, Evan J. Lipson, Mac Cheever, Robert A. Anders, Paul T. Nghiem, Suzanne L. Topalian, and Janis M. Taube
- Subjects
PD-1 ,PD-L1 ,Merkel cell ,Multispectral immunofluorescence ,Pembrolizumab ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background We recently reported a 56% objective response rate in patients with advanced Merkel cell carcinoma (MCC) receiving pembrolizumab. However, a biomarker predicting clinical response was not identified. Methods Pretreatment FFPE tumor specimens (n = 26) were stained for CD8, PD-L1, and PD-1 by immunohistochemistry/immunofluorescence (IHC/IF), and the density and distribution of positive cells was quantified to determine the associations with anti-PD-1 response. Multiplex IF was used to test a separate cohort of MCC archival specimens (n = 16), to identify cell types expressing PD-1. Results Tumors from patients who responded to anti-PD-1 showed higher densities of PD-1+ and PD-L1+ cells when compared to non-responders (median cells/mm2, 70.7 vs. 6.7, p = 0.03; and 855.4 vs. 245.0, p = 0.02, respectively). There was no significant association of CD8+ cell density with clinical response. Quantification of PD-1+ cells located within 20 μm of a PD-L1+ cell showed that PD-1/PD-L1 proximity was associated with clinical response (p = 0.03), but CD8/PD-L1 proximity was not. CD4+ and CD8+ cells in the TME expressed similar amounts of PD-1. Conclusions While the binomial presence or absence of PD-L1 expression in the TME was not sufficient to predict response to anti-PD-1 in patients with MCC, we show that quantitative assessments of PD-1+ and PD-L1+ cell densities as well as the geographic interactions between these two cell populations correlate with clinical response. Cell types expressing PD-1 in the TME include CD8+ T-cells, CD4+ T-cells, Tregs, and CD20+ B-cells, supporting the notion that multiple cell types may potentiate tumor regression following PD-1 blockade.
- Published
- 2018
- Full Text
- View/download PDF
7. Data from Dynamics of Tumor and Immune Responses during Immune Checkpoint Blockade in Non–Small Cell Lung Cancer
- Author
-
Victor E. Velculescu, Julie R. Brahmer, Matthew D. Hellmann, Jamie E. Chaft, Drew M. Pardoll, Rachel Karchin, Robert B. Scharpf, Janis Taube, Jennifer L. Sauter, James M. Isbell, Malcolm V. Brock, Edward Gabrielson, Qing Kay Li, Peter Illei, Franco Verde, Christos Georgiades, Josephine Feliciano, Benjamin Levy, Christine L. Hann, Doreen N. Palsgrove, Lamia Rhymee, Tricia R. Cottrell, Kellie N. Smith, Vilmos Adleff, Alessandro Leal, Jillian Phallen, Samuel Rosner, Daniel C. Bruhm, I.K. Ashok Sivakumar, Kristen Marrone, Jarushka Naidoo, Carolyn Hruban, Noushin Niknafs, James R. White, Patrick M. Forde, and Valsamo Anagnostou
- Abstract
Despite the initial successes of immunotherapy, there is an urgent clinical need for molecular assays that identify patients more likely to respond. Here, we report that ultrasensitive measures of circulating tumor DNA (ctDNA) and T-cell expansion can be used to assess responses to immune checkpoint blockade in metastatic lung cancer patients (N = 24). Patients with clinical response to therapy had a complete reduction in ctDNA levels after initiation of therapy, whereas nonresponders had no significant changes or an increase in ctDNA levels. Patients with initial response followed by acquired resistance to therapy had an initial drop followed by recrudescence in ctDNA levels. Patients without a molecular response had shorter progression-free and overall survival compared with molecular responders [5.2 vs. 14.5 and 8.4 vs. 18.7 months; HR 5.36; 95% confidence interval (CI), 1.57–18.35; P = 0.007 and HR 6.91; 95% CI, 1.37–34.97; P = 0.02, respectively], which was detected on average 8.7 weeks earlier and was more predictive of clinical benefit than CT imaging. Expansion of T cells, measured through increases of T-cell receptor productive frequencies, mirrored ctDNA reduction in response to therapy. We validated this approach in an independent cohort of patients with early-stage non–small cell lung cancer (N = 14), where the therapeutic effect was measured by pathologic assessment of residual tumor after anti-PD1 therapy. Consistent with our initial findings, early ctDNA dynamics predicted pathologic response to immune checkpoint blockade. These analyses provide an approach for rapid determination of therapeutic outcomes for patients treated with immune checkpoint inhibitors and have important implications for the development of personalized immune targeted strategies.Significance: Rapid and sensitive detection of circulating tumor DNA dynamic changes and T-cell expansion can be used to guide immune targeted therapy for patients with lung cancer.See related commentary by Zou and Meyerson, p. 1038
- Published
- 2023
8. Table S2 from Compartmental Analysis of T-cell Clonal Dynamics as a Function of Pathologic Response to Neoadjuvant PD-1 Blockade in Resectable Non–Small Cell Lung Cancer
- Author
-
Kellie N. Smith, Hongkai Ji, Drew M. Pardoll, Patrick M. Forde, Matthew D. Hellmann, Franck Housseau, Julie R. Brahmer, Victor E. Velculescu, Janis M. Taube, Edward Gabrielson, Jarushka Naidoo, Kristen A. Marrone, David R. Jones, Jinny S. Ha, Ni Zhao, John-William Sidhom, Mohsen Abu-Akeel, Richard L. Blosser, Ada J. Tam, Joshua E. Reuss, Jamie E. Chaft, Taha Merghoub, Haidan Guo, Prerna Suri, Hok Yee Chan, Tricia R. Cottrell, Valsamo Anagnostou, Margueritta El Asmar, Justina X. Caushi, Zhicheng Ji, and Jiajia Zhang
- Abstract
Correlative assays performed on each sample
- Published
- 2023
9. Data from Compartmental Analysis of T-cell Clonal Dynamics as a Function of Pathologic Response to Neoadjuvant PD-1 Blockade in Resectable Non–Small Cell Lung Cancer
- Author
-
Kellie N. Smith, Hongkai Ji, Drew M. Pardoll, Patrick M. Forde, Matthew D. Hellmann, Franck Housseau, Julie R. Brahmer, Victor E. Velculescu, Janis M. Taube, Edward Gabrielson, Jarushka Naidoo, Kristen A. Marrone, David R. Jones, Jinny S. Ha, Ni Zhao, John-William Sidhom, Mohsen Abu-Akeel, Richard L. Blosser, Ada J. Tam, Joshua E. Reuss, Jamie E. Chaft, Taha Merghoub, Haidan Guo, Prerna Suri, Hok Yee Chan, Tricia R. Cottrell, Valsamo Anagnostou, Margueritta El Asmar, Justina X. Caushi, Zhicheng Ji, and Jiajia Zhang
- Abstract
Purpose:Neoadjuvant PD-1 blockade is a promising treatment for resectable non–small cell lung cancer (NSCLC), yet immunologic mechanisms contributing to tumor regression and biomarkers of response are unknown. Using paired tumor/blood samples from a phase II clinical trial (NCT02259621), we explored whether the peripheral T-cell clonotypic dynamics can serve as a biomarker for response to neoadjuvant PD-1 blockade.Experimental Design:T-cell receptor (TCR) sequencing was performed on serial peripheral blood, tumor, and normal lung samples from resectable NSCLC patients treated with neoadjuvant PD-1 blockade. We explored the temporal dynamics of the T-cell repertoire in the peripheral and tumoral compartments in response to neoadjuvant PD-1 blockade by using the TCR as a molecular barcode.Results:Higher intratumoral TCR clonality was associated with reduced percent residual tumor at the time of surgery, and the TCR repertoire of tumors with major pathologic response (MPR; Conclusions:Our study suggests that exchange of T-cell clones between tumor and blood represents a key correlate of pathologic response to neoadjuvant immunotherapy and shows that the periphery may be a previously underappreciated originating compartment for effective antitumor immunity.See related commentary by Henick, p. 1205
- Published
- 2023
10. Supplementary Table S3 from Transcriptional Mechanisms of Resistance to Anti–PD-1 Therapy
- Author
-
Suzanne L. Topalian, Christine A. Iacobuzio-Donahue, David B. Solit, Barry S. Taylor, Drew M. Pardoll, Ralph H. Hruban, Leslie Cope, Timothy A. Chan, Nadeem Riaz, Vladimir Makarov, Alexander Favorov, Zachary A. Kohutek, Tricia R. Cottrell, Genevieve J. Kaunitz, Jinshui Fan, Alan E. Berger, Tracee L. McMiller, Janis M. Taube, Evan J. Lipson, Alvin Makohon-Moore, and Maria L. Ascierto
- Abstract
140 Illumina probes corresponding to138 unique genes differentially expressed in regressing (R) vs. progressing (P) cutaneous metastases based on whole genome microarray analysis (fold change magnitude {greater than or equal to}1.7, p value {less than or equal to}0.05 )
- Published
- 2023
11. Data from PD-L1 Expression in Melanoma: A Quantitative Immunohistochemical Antibody Comparison
- Author
-
Janis M. Taube, Robert A. Anders, Evan J. Lipson, Toby C. Cornish, Karen B. Bleich, Aleksandra Ogurtsova, Haiying Xu, Jessica Esandrio, Sneha Berry, Tricia R. Cottrell, Genevieve J. Kaunitz, Peter L. Nguyen, and Joel C. Sunshine
- Abstract
Purpose: PD-L1 expression in the pretreatment tumor microenvironment enriches for response to anti-PD-1/PD-L1 therapies. The purpose of this study was to quantitatively compare the performance of five monoclonal anti-PD-L1 antibodies used in recent landmark publications.Experimental Design: PD-L1 IHC was performed on 34 formalin-fixed paraffin-embedded archival melanoma samples using the 5H1, SP142, 28-8, 22C3, and SP263 clones. The percentage of total cells (including melanocytes and immune cells) demonstrating cell surface PD-L1 staining, as well as intensity measurements/H-scores, were assessed for each melanoma specimen using a computer-assisted platform. Staining properties were compared between antibodies.Results: Strong correlations were observed between the percentage of PD-L1(+) cells across all clones studied (R2 = 0.81–0.96). When present, discordant results were attributable to geographic heterogeneity of the melanoma tissue section rather than differences in PD-L1 antibody staining characteristics. PD-L1 intensity/H-scores strongly correlated with percentage of PD-L1(+) cells (R2 > 0.78, all clones).Conclusions: The 5H1, SP142, 28-8, 22C3, and SP263 clones all demonstrated similar performance characteristics when used in a standardized IHC assay on melanoma specimens. Reported differences in PD-L1 IHC assays using these antibodies are thus most likely due to assay characteristics beyond the antibody itself. Our findings also argue against the inclusion of an intensity/H-score in chromogenic PD-L1 IHC assays. Clin Cancer Res; 23(16); 4938–44. ©2017 AACR.
- Published
- 2023
12. Supplementary Data from PD-L1 Expression in Melanoma: A Quantitative Immunohistochemical Antibody Comparison
- Author
-
Janis M. Taube, Robert A. Anders, Evan J. Lipson, Toby C. Cornish, Karen B. Bleich, Aleksandra Ogurtsova, Haiying Xu, Jessica Esandrio, Sneha Berry, Tricia R. Cottrell, Genevieve J. Kaunitz, Peter L. Nguyen, and Joel C. Sunshine
- Abstract
Supplemental Figures S1 Figure S2, Figure S3 and Table S1 and Table S2
- Published
- 2023
13. Supplementary Figures from Compartmental Analysis of T-cell Clonal Dynamics as a Function of Pathologic Response to Neoadjuvant PD-1 Blockade in Resectable Non–Small Cell Lung Cancer
- Author
-
Kellie N. Smith, Hongkai Ji, Drew M. Pardoll, Patrick M. Forde, Matthew D. Hellmann, Franck Housseau, Julie R. Brahmer, Victor E. Velculescu, Janis M. Taube, Edward Gabrielson, Jarushka Naidoo, Kristen A. Marrone, David R. Jones, Jinny S. Ha, Ni Zhao, John-William Sidhom, Mohsen Abu-Akeel, Richard L. Blosser, Ada J. Tam, Joshua E. Reuss, Jamie E. Chaft, Taha Merghoub, Haidan Guo, Prerna Suri, Hok Yee Chan, Tricia R. Cottrell, Valsamo Anagnostou, Margueritta El Asmar, Justina X. Caushi, Zhicheng Ji, and Jiajia Zhang
- Abstract
Figures S1-S8
- Published
- 2023
14. Supplementary Tables S1-S18 from Dynamics of Tumor and Immune Responses during Immune Checkpoint Blockade in Non–Small Cell Lung Cancer
- Author
-
Victor E. Velculescu, Julie R. Brahmer, Matthew D. Hellmann, Jamie E. Chaft, Drew M. Pardoll, Rachel Karchin, Robert B. Scharpf, Janis Taube, Jennifer L. Sauter, James M. Isbell, Malcolm V. Brock, Edward Gabrielson, Qing Kay Li, Peter Illei, Franco Verde, Christos Georgiades, Josephine Feliciano, Benjamin Levy, Christine L. Hann, Doreen N. Palsgrove, Lamia Rhymee, Tricia R. Cottrell, Kellie N. Smith, Vilmos Adleff, Alessandro Leal, Jillian Phallen, Samuel Rosner, Daniel C. Bruhm, I.K. Ashok Sivakumar, Kristen Marrone, Jarushka Naidoo, Carolyn Hruban, Noushin Niknafs, James R. White, Patrick M. Forde, and Valsamo Anagnostou
- Abstract
Supplementary Table S1. Summary of patient characteristics Supplementary Table S2. Summary of plasma samples analyzed Supplementary Table S3. Summary of peripheral blood lymphocytes and tumor infiltrating lymphocytes samples analyzed Supplementary Table S4. Genes analyzed by TEC-Seq Supplementary Table S5. Summary of TEC-Seq Characteristics Supplementary Table S6. Somatic sequence alterations detected in cfDNA Supplementary Table S7. Mutation Cellularity Analysis for tumor-specific cfDNA variants Supplementary Table S8. TCR-beta sequencing analysis Supplementary Table S9. Differentially abundant clones for CGLU111 Supplementary Table S10. Differentially abundant clones for CGLU117 Supplementary Table S11. Differentially abundant clones for CGLU127 Supplementary Table S12. Differentially abundant clones for CGLU212 Supplementary Table S13. Differentially abundant clones for CGLU135 Supplementary Table S14. Differentially abundant clones for CGLU161 Supplementary Table S15. Differentially abundant clones for CGLU159 Supplementary Table S16. Differentially abundant clones for CGLU162 Supplementary Table S17. Differentially abundant clones for CGLU203 Supplementary Table S18. Differentially abundant clones for CGLU243
- Published
- 2023
15. Data from Transcriptional Mechanisms of Resistance to Anti–PD-1 Therapy
- Author
-
Suzanne L. Topalian, Christine A. Iacobuzio-Donahue, David B. Solit, Barry S. Taylor, Drew M. Pardoll, Ralph H. Hruban, Leslie Cope, Timothy A. Chan, Nadeem Riaz, Vladimir Makarov, Alexander Favorov, Zachary A. Kohutek, Tricia R. Cottrell, Genevieve J. Kaunitz, Jinshui Fan, Alan E. Berger, Tracee L. McMiller, Janis M. Taube, Evan J. Lipson, Alvin Makohon-Moore, and Maria L. Ascierto
- Abstract
Purpose: To explore factors associated with response and resistance to anti–PD-1 therapy, we analyzed multiple disease sites at autopsy in a patient with widely metastatic melanoma who had a heterogeneous response.Materials and Methods: Twenty-six melanoma specimens (four premortem, 22 postmortem) were subjected to whole exome sequencing. Candidate immunologic markers and gene expression were assessed in 10 cutaneous metastases showing response or progression during therapy.Results: The melanoma was driven by biallelic inactivation of NF1. All lesions had highly concordant mutational profiles and copy number alterations, indicating linear clonal evolution. Expression of candidate immunologic markers was similar in responding and progressing lesions. However, progressing cutaneous metastases were associated with overexpression of genes associated with extracellular matrix and neutrophil function.Conclusions: Although mutational and immunologic differences have been proposed as the primary determinants of heterogeneous response/resistance to targeted therapies and immunotherapies, respectively, differential lesional gene expression profiles may also dictate anti–PD-1 outcomes. Clin Cancer Res; 23(12); 3168–80. ©2017 AACR.See related commentary by Wilmott et al., p. 2921
- Published
- 2023
16. Supplemental Appendix from Pan-Tumor Pathologic Scoring of Response to PD-(L)1 Blockade
- Author
-
Janis M. Taube, Suzanne L. Topalian, Drew M. Pardoll, Elizabeth M. Jaffee, Hao Wang, Josephine Feliciano, Mark Yarchoan, Mohamad E. Allaf, Elizabeth D. Thompson, Ashley Cimino-Mathews, Robert A. Anders, Patrick M. Forde, Tricia R. Cottrell, Evan J. Lipson, and Julie E. Stein
- Abstract
Supplemental Appendix (Figures S1 and S2, Table S1)
- Published
- 2023
17. Data from Pan-Tumor Pathologic Scoring of Response to PD-(L)1 Blockade
- Author
-
Janis M. Taube, Suzanne L. Topalian, Drew M. Pardoll, Elizabeth M. Jaffee, Hao Wang, Josephine Feliciano, Mark Yarchoan, Mohamad E. Allaf, Elizabeth D. Thompson, Ashley Cimino-Mathews, Robert A. Anders, Patrick M. Forde, Tricia R. Cottrell, Evan J. Lipson, and Julie E. Stein
- Abstract
Purpose:Pathologic response assessment of tumor specimens from patients receiving systemic treatment provides an early indication of therapeutic efficacy and predicts long-term survival. Grading systems for pathologic response were first developed for chemotherapy in select tumor types. Immunotherapeutic agents have a mechanism of action distinct from chemotherapy and are being used across a broad array of tumor types. A standardized, universal scoring system for pathologic response that encompasses features characteristic for immunotherapy and spans tumor types is needed.Experimental Design:Hematoxylin and eosin–stained slides from neoadjuvant surgical resections and on-treatment biopsies were assessed for features of immune-related pathologic response (irPR). A total of 258 specimens from patients with 11 tumor types as part of ongoing clinical trials for anti-PD-(L)1 were evaluated. An additional 98 specimens from patients receiving anti-PD-(L)1 in combination with other treatments were also reviewed, including those from three additional tumor types.Results:Common irPR features (immune activation, cell death, tissue repair, and regression bed) were present in all tumor types reviewed, including melanoma, non–small cell lung, head and neck squamous cell, Merkel cell, and renal cell carcinoma, among others. Features were consistent across primary tumors, lymph nodes, and distant metastases. Specimens from patients treated with anti-PD-(L)1 in combination with another agent also exhibited irPR features.Conclusions:irPR features are consistent across tumor types and treatment settings. Standardized, pan-tumor irPR criteria (irPRC) are defined and associated specimen-handling considerations are described. Future, prospective studies are merited to validate irPRC in larger datasets and to associate pathologic features with long-term patient outcomes.
- Published
- 2023
18. Supplementary Methods from Transcriptional Mechanisms of Resistance to Anti–PD-1 Therapy
- Author
-
Suzanne L. Topalian, Christine A. Iacobuzio-Donahue, David B. Solit, Barry S. Taylor, Drew M. Pardoll, Ralph H. Hruban, Leslie Cope, Timothy A. Chan, Nadeem Riaz, Vladimir Makarov, Alexander Favorov, Zachary A. Kohutek, Tricia R. Cottrell, Genevieve J. Kaunitz, Jinshui Fan, Alan E. Berger, Tracee L. McMiller, Janis M. Taube, Evan J. Lipson, Alvin Makohon-Moore, and Maria L. Ascierto
- Abstract
Supplementary methods
- Published
- 2023
19. Supplementary Figures S1-S14 from Dynamics of Tumor and Immune Responses during Immune Checkpoint Blockade in Non–Small Cell Lung Cancer
- Author
-
Victor E. Velculescu, Julie R. Brahmer, Matthew D. Hellmann, Jamie E. Chaft, Drew M. Pardoll, Rachel Karchin, Robert B. Scharpf, Janis Taube, Jennifer L. Sauter, James M. Isbell, Malcolm V. Brock, Edward Gabrielson, Qing Kay Li, Peter Illei, Franco Verde, Christos Georgiades, Josephine Feliciano, Benjamin Levy, Christine L. Hann, Doreen N. Palsgrove, Lamia Rhymee, Tricia R. Cottrell, Kellie N. Smith, Vilmos Adleff, Alessandro Leal, Jillian Phallen, Samuel Rosner, Daniel C. Bruhm, I.K. Ashok Sivakumar, Kristen Marrone, Jarushka Naidoo, Carolyn Hruban, Noushin Niknafs, James R. White, Patrick M. Forde, and Valsamo Anagnostou
- Abstract
Supplementary Figure S1. ctDNA clonal dynamics during anti-PD1 for patients with a clinical response. Supplementary Figure S2. ctDNA clonal dynamics during anti-PD1 for patients with molecular primary resistance. Supplementary Figure S3. ctDNA molecular responses precede radiologic responses. Supplementary Figure S4. Molecular responses correlate with a favorable prognosis. Supplementary Figure S5. Duration of molecular response correlates with progression-free and overall survival. Supplementary Figure S6. Disease prognostication by radiologic imaging at first assessment. Supplementary Figure S7. Differential clinical outcome by molecular responses in patients with radiologically stable disease. Supplementary Figure S8. Disease prognostication by tumor mutation burden. Supplementary Figure S9. ctDNA molecular response more accurately distinguish clinical responders from non-responders compared to clonal tumor mutation burden. Supplementary Figure S10. Molecular responses mirror pathologic responses to anti-PD1 therapy in early stage operable NSCLC. Supplementary Figure S11. TCR clonal dynamics during response and acquired resistance to anti-PD1. Supplementary Figure S12. TCR clonal dynamics for patients with clinical primary resistance. Supplementary Figure S13. Differential VJ gene usage for a patient with clinical response compared to a patient with clinical primary resistance. Supplementary Figure S14. Dynamic changes in pro-inflammatory and immunosuppressive cytokines in early stage NSCLC patients with differential responses to anti-PD1 therapy.
- Published
- 2023
20. Correction to: persistent mutant oncogene specific T cells in two patients benefitting from anti-PD-1
- Author
-
Kellie N. Smith, Nicolas J. Llosa, Tricia R. Cottrell, Nicholas Siegel, Hongni Fan, Prerna Suri, Hok Yee Chan, Haidan Guo, Teniola Oke, Anas H. Awan, Franco Verde, Ludmila Danilova, Valsamo Anagnostou, Ada J. Tam, Brandon S. Luber, Bjarne R. Bartlett, Laveet K. Aulakh, John-William Sidhom, Qingfeng Zhu, Cynthia L. Sears, Leslie Cope, William H. Sharfman, Elizabeth D. Thompson, Joanne Riemer, Kristen A. Marrone, Jarushka Naidoo, Victor E. Velculescu, Patrick M. Forde, Bert Vogelstein, Kenneth W. Kinzler, Nickolas Papadopoulos, Jennifer N. Durham, Hao Wang, Dung T. Le, Sune Justesen, Janis M. Taube, Luis A. Diaz Jr, Julie R. Brahmer, Drew M. Pardoll, Robert A. Anders, and Franck Housseau
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2019
- Full Text
- View/download PDF
21. Immune cell subsets in interface cutaneous immune-related adverse events associated with anti-PD-1 therapy resemble acute graft versus host disease more than lichen planus
- Author
-
Guillermo E. Almodovar Cruz, Genevieve Kaunitz, Julie E. Stein, Inbal Sander, Travis Hollmann, Tricia R. Cottrell, Janis M. Taube, and Joel C. Sunshine
- Subjects
Histology ,T-Lymphocytes ,Lichen Planus ,Graft vs Host Disease ,Humans ,Dermatology ,Immunohistochemistry ,Pathology and Forensic Medicine ,Skin - Abstract
Checkpoint immunotherapy is frequently associated with cutaneous immune-related adverse events (cirAEs), and among those, the most common subtype shows interface reaction patterns that have been likened to lichen planus (LP); however, cutaneous acute graft versus host disease (aGVHD) may be a closer histopathologic comparator. We used quantitative pathology to compare the immunologic composition of anti-PD-1-associated interface reactions to LP and aGVHD to assess for similarities and differences between these cutaneous eruptions.Immunohistochemistry for CD4, CD8, CD68, PD-1, and PD-L1 was performed on formalin-fixed paraffin-embedded tissue from patients with anti-PD-1 interface cirAEs (n = 4), LP (n = 9), or aGVHD (n = 5). Densities of immune cell subsets expressing each marker were quantified using the HALO image analysis immune cell module. Plasma cell and eosinophil density were quantified on routine HE slides.Specimens from patients with anti-PD-1 interface cirAEs showed equivalent total cell densities and immune cell composition to those with aGVHD. Patients with LP showed higher total immune cell infiltration, higher absolute T-cell densities, increased CD8 proportion, and reduced histiocytic component. The cases with the highest plasma cell counts were all anti-PD-1 interface cirAEs and aGVHD.The composition of immune cell subsets in anti-PD-1 interface cirAEs more closely resembles the immune response seen in aGVHD than LP within our cohort. This warrants a closer look via advanced analytics and may have implications for shared pathogenesis and potential treatment options.
- Published
- 2022
22. Expression of the Autoantigen Topoisomerase‐1 is Enriched in the Lung Tissues of Patients With Autoimmune Interstitial Lung Disease: A Case Control Study
- Author
-
Livia Casciola-Rosen, Frederic B. Askin, Marc K. Halushka, Zsuzsanna H. McMahan, and Tricia R. Cottrell
- Subjects
Pathology ,medicine.medical_specialty ,lcsh:Diseases of the musculoskeletal system ,Lung ,biology ,business.industry ,Brief Report ,Interstitial lung disease ,Autoantibody ,respiratory system ,medicine.disease ,Scleroderma ,Immune system ,medicine.anatomical_structure ,Rheumatology ,Antigen ,biology.protein ,Medicine ,lcsh:RC925-935 ,Antibody ,business ,CD8 - Abstract
Background Among the autoimmune rheumatic diseases, it is striking that autoantibodies targeting ubiquitously expressed proteins (eg, topoisomerase-1) associate with specific clinical complications (eg, interstitial lung disease [ILD]). It has been proposed that enriched antigen expression in inflamed target tissue may play a role in focusing the autoimmune response. We sought to determine whether topoisomerase-1 expression is enriched in lungs from patients with autoimmune/inflammatory diseases relative to normal lung. Methods We used a 99-core lung tissue microarray (TMA) containing lung tissue from 40 patients with autoimmune inflammatory ILD (cases) and 46 control subjects with normal lungs. We stained the TMA with antibodies to compare topoisomerase-1 and CD8 expression between patients and control subjects and evaluated whether expression is enriched in specific cell types. Staining was analyzed, and statistical comparisons were performed. Results Cases were more likely to have global topoisomerase-1 expression (53% vs 21%; P = 0.003), specifically in pneumocytes (47% vs 16%; P = 0.003) and stromal/immune cells (32% vs 5%; P = 0.002) compared with control subjects. CD8 cell density (223 cells/mm2 vs 102 cells/mm2 ; P = 0.018) was significantly higher in topoisomerase-1-positive lung tissues compared with topoisomerase-1-negative lung tissues. Interestingly, topoisomerase-1 expression was significantly more common in scleroderma compared with normal lung (67% vs 21%; P = 0.036) and was present more frequently in pneumocytes in these patients (67% vs 16%; P = 0.018). Conclusions Pulmonary expression of topoisomerase-1 is increased in the setting of autoimmune ILD relative to normal lung, specifically in pneumocytes. This may contribute to the amplification of pulmonary disease in patients with scleroderma with a loss of tolerance to topoisomerase-1.
- Published
- 2020
23. Compartmental Analysis of T-cell Clonal Dynamics as a Function of Pathologic Response to Neoadjuvant PD-1 Blockade in Resectable Non–Small Cell Lung Cancer
- Author
-
Justina X. Caushi, Haidan Guo, Tricia R. Cottrell, Matthew D. Hellmann, Drew M. Pardoll, Ni Zhao, John-William Sidhom, Margueritta El Asmar, Hok Yee Chan, Patrick M. Forde, Richard L. Blosser, Zhicheng Ji, Valsamo Anagnostou, Jiajia Zhang, Jamie E. Chaft, David R. Jones, Edward Gabrielson, Hongkai Ji, Taha Merghoub, Prerna Suri, Victor E. Velculescu, Joshua E. Reuss, Kristen A. Marrone, Jarushka Naidoo, Jinny Ha, Kellie N. Smith, Janis M. Taube, Ada J. Tam, Mohsen Abu-Akeel, Julie R. Brahmer, and Franck Housseau
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,03 medical and health sciences ,0302 clinical medicine ,Major Pathologic Response ,Carcinoma, Non-Small-Cell Lung ,Carcinoma ,medicine ,Humans ,Lung cancer ,Neoadjuvant therapy ,business.industry ,T-cell receptor ,Immunotherapy ,medicine.disease ,Neoadjuvant Therapy ,Blockade ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Biomarker (medicine) ,business - Abstract
Purpose:Neoadjuvant PD-1 blockade is a promising treatment for resectable non–small cell lung cancer (NSCLC), yet immunologic mechanisms contributing to tumor regression and biomarkers of response are unknown. Using paired tumor/blood samples from a phase II clinical trial (NCT02259621), we explored whether the peripheral T-cell clonotypic dynamics can serve as a biomarker for response to neoadjuvant PD-1 blockade.Experimental Design:T-cell receptor (TCR) sequencing was performed on serial peripheral blood, tumor, and normal lung samples from resectable NSCLC patients treated with neoadjuvant PD-1 blockade. We explored the temporal dynamics of the T-cell repertoire in the peripheral and tumoral compartments in response to neoadjuvant PD-1 blockade by using the TCR as a molecular barcode.Results:Higher intratumoral TCR clonality was associated with reduced percent residual tumor at the time of surgery, and the TCR repertoire of tumors with major pathologic response (MPR; Conclusions:Our study suggests that exchange of T-cell clones between tumor and blood represents a key correlate of pathologic response to neoadjuvant immunotherapy and shows that the periphery may be a previously underappreciated originating compartment for effective antitumor immunity.See related commentary by Henick, p. 1205
- Published
- 2020
24. Pan-Tumor Pathologic Scoring of Response to PD-(L)1 Blockade
- Author
-
Tricia R. Cottrell, Patrick M. Forde, Elizabeth M. Jaffee, Janis M. Taube, Josephine Feliciano, Mohamad E. Allaf, Julie E. Stein, Evan J. Lipson, Robert A. Anders, Ashley Cimino-Mathews, Mark Yarchoan, Elizabeth D. Thompson, Drew M. Pardoll, Hao Wang, and Suzanne L. Topalian
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,H&E stain ,B7-H1 Antigen ,Article ,03 medical and health sciences ,0302 clinical medicine ,Renal cell carcinoma ,Neoplasms ,Internal medicine ,medicine ,Humans ,Grading (tumors) ,Response Evaluation Criteria in Solid Tumors ,Chemotherapy ,Pathology, Clinical ,business.industry ,Melanoma ,Immunotherapy ,medicine.disease ,Clinical trial ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Research Design ,030220 oncology & carcinogenesis ,business ,Merkel cell - Abstract
Purpose: Pathologic response assessment of tumor specimens from patients receiving systemic treatment provides an early indication of therapeutic efficacy and predicts long-term survival. Grading systems for pathologic response were first developed for chemotherapy in select tumor types. Immunotherapeutic agents have a mechanism of action distinct from chemotherapy and are being used across a broad array of tumor types. A standardized, universal scoring system for pathologic response that encompasses features characteristic for immunotherapy and spans tumor types is needed. Experimental Design: Hematoxylin and eosin–stained slides from neoadjuvant surgical resections and on-treatment biopsies were assessed for features of immune-related pathologic response (irPR). A total of 258 specimens from patients with 11 tumor types as part of ongoing clinical trials for anti-PD-(L)1 were evaluated. An additional 98 specimens from patients receiving anti-PD-(L)1 in combination with other treatments were also reviewed, including those from three additional tumor types. Results: Common irPR features (immune activation, cell death, tissue repair, and regression bed) were present in all tumor types reviewed, including melanoma, non–small cell lung, head and neck squamous cell, Merkel cell, and renal cell carcinoma, among others. Features were consistent across primary tumors, lymph nodes, and distant metastases. Specimens from patients treated with anti-PD-(L)1 in combination with another agent also exhibited irPR features. Conclusions: irPR features are consistent across tumor types and treatment settings. Standardized, pan-tumor irPR criteria (irPRC) are defined and associated specimen-handling considerations are described. Future, prospective studies are merited to validate irPRC in larger datasets and to associate pathologic features with long-term patient outcomes.
- Published
- 2020
25. Evaluating T-cell cross-reactivity between tumors and immune-related adverse events with TCR sequencing: pitfalls in interpretations of functional relevance
- Author
-
Genevieve J. Kaunitz, Janis M. Taube, Boyang Zhang, Poromendro Burman, Tricia R. Cottrell, Hongkai Ji, Jody E. Hooper, Mohamad E. Allaf, Franco Verde, Hans J. Hammers, Jiajia Zhang, Kellie N. Smith, and Hok Yee Chan
- Subjects
0301 basic medicine ,Cancer Research ,Drug-Related Side Effects and Adverse Reactions ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Immunology ,Receptors, Antigen, T-Cell ,receptors ,biostatistics ,Case Report ,immunologic techniques ,Cross Reactions ,medicine.disease_cause ,Bioinformatics ,Cross-reactivity ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Neoplasms ,Immunology and Allergy ,Medicine ,Humans ,immunologic ,RC254-282 ,Aged ,Pharmacology ,business.industry ,T-cell receptor ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,Immune checkpoint ,Blockade ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,business - Abstract
T-cell receptor sequencing (TCRseq) enables tracking of T-cell clonotypes recognizing the same antigen over time and across biological compartments. TCRseq has been used to test if cross-reactive antitumor T cells are responsible for development of immune-related adverse events (irAEs) following immune checkpoint blockade. Prior studies have interpreted T-cell clones shared among the tumor and irAE as evidence supporting this, but interpretations of these findings are challenging, given the constraints of TCRseq. Here we capitalize on a rare opportunity to understand the impact of potential confounders, such as sample size, tissue compartment, and collection batch/timepoint, on the relative proportion of shared T-cell clones between an irAE and tumor specimens. TCRseq was performed on tumor-involved and -uninvolved tissues, including an irAE, that were obtained throughout disease progression and at the time of rapid autopsy from a patient with renal cell carcinoma treated with programmed death-1 (PD-1) blockade. Our analyses show significant effects of these confounders on our ability to understand T-cell receptor overlap, and we present mitigation strategies and study design recommendations to reduce these errors. Implementation of these strategies will enable more rigorous TCRseq-based studies of immune responses in human tissues, particularly as they relate to antitumor T-cell cross-reactivity in irAEs following checkpoint blockade.
- Published
- 2021
26. Major pathologic response on biopsy (MPRbx) in patients with advanced melanoma treated with anti-PD-1: evidence for an early, on-therapy biomarker of response
- Author
-
Thomas F. Gajewski, Evan J. Lipson, Tricia R. Cottrell, F.S. Hodi, Julie E. Stein, Megan Wind-Rotolo, Janis M. Taube, Shailender Bhatia, Abha Soni, Robin Edwards, Liudmila V Danilova, William H. Sharfman, and W J Urba
- Subjects
Adult ,Male ,0301 basic medicine ,Oncology ,medicine.medical_specialty ,Neoplasm, Residual ,Skin Neoplasms ,Necrosis ,Biopsy ,Programmed Cell Death 1 Receptor ,H&E stain ,Kaplan-Meier Estimate ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,Stable Disease ,Major Pathologic Response ,Fibrosis ,Internal medicine ,Biomarkers, Tumor ,medicine ,Humans ,Prospective Studies ,Melanoma ,Response Evaluation Criteria in Solid Tumors ,Aged ,Skin ,medicine.diagnostic_test ,business.industry ,Original Articles ,Hematology ,Middle Aged ,medicine.disease ,Ipilimumab ,Nivolumab ,030104 developmental biology ,030220 oncology & carcinogenesis ,Biomarker (medicine) ,Female ,medicine.symptom ,business - Abstract
BACKGROUND: With increasing anti-PD-1 therapy use in patients with melanoma and other tumor types, there is interest in developing early on-treatment biomarkers that correlate with long-term patient outcome. An understanding of the pathologic features of immune-mediated tumor regression is key in this endeavor. MATERIALS AND METHODS: Histologic features of immune-related pathologic response (irPR) following anti-PD-1 therapy were identified on hematoxylin and eosin (H&E)-stained slides in a discovery cohort of pre- and on-treatment specimens from n = 16 patients with advanced melanoma. These features were used to generate an irPR score [from 0 = no irPR features to 3 = major pathologic response on biopsy (MPR(bx), ≤10% residual viable tumor)]. This scoring system was then tested for an association with objective response by RECIST1.1 and overall survival in a prospectively collected validation cohort of pre- and on-treatment biopsies (n = 51 on-treatment at 4-week timepoint) from melanoma patients enrolled on the nivolumab monotherapy arm of CA209-038 (NCT01621490). RESULTS: Specimens from responders in the discovery cohort had features of immune-activation (moderate–high TIL densities, plasma cells) and wound-healing/tissue repair (neovascularization, proliferative fibrosis) compared to nonresponders, (P ≤ 0.021, for each feature). In the validation cohort, increasing irPR score associated with objective response (P = 0.009) and MPR(bx) associated with increased overall survival (n = 51; HR 0.13; 95%CI, 0.054–0.31, P = 0.015). Neither tumoral necrosis nor pretreatment histologic features were associated with response. Eight of 16 (50%) of patients with stable disease showed irPR features, two of which were MPR(bx), indicating a disconnect between pathologic and radiographic features at the 4-week on-therapy timepoint for some patients. CONCLUSIONS: Features of immune-mediated tumor regression on routine H&E-stained biopsy slides from patients with advanced melanoma correlate with objective response to anti-PD-1 and overall survival. An on-therapy biopsy may be particularly clinically useful for informing treatment decisions in patients with radiographic stable disease. This approach is inexpensive, straightforward, and widely available.
- Published
- 2019
27. Dynamics of Tumor and Immune Responses during Immune Checkpoint Blockade in Non–Small Cell Lung Cancer
- Author
-
Malcolm V. Brock, Benjamin Levy, Jillian Phallen, Matthew D. Hellmann, Victor E. Velculescu, Doreen N. Palsgrove, I K Ashok Sivakumar, Edward Gabrielson, Qing Kay Li, Christine L. Hann, Lamia Rhymee, Valsamo Anagnostou, Peter B. Illei, Patrick M. Forde, Kristen A. Marrone, Alessandro Leal, Josephine Feliciano, Jarushka Naidoo, Daniel C. Bruhm, Franco Verde, James M. Isbell, James R. White, Carolyn Hruban, Janis M. Taube, Tricia R. Cottrell, Robert B. Scharpf, Vilmos Adleff, Drew M. Pardoll, Julie R. Brahmer, Christos S. Georgiades, Rachel Karchin, Noushin Niknafs, Jennifer L. Sauter, Jamie E. Chaft, Kellie N. Smith, and Samuel Rosner
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Immunotherapy ,medicine.disease ,Article ,Immune checkpoint ,Blockade ,Targeted therapy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,030220 oncology & carcinogenesis ,Molecular Response ,Internal medicine ,medicine ,Lung cancer ,business ,Survival rate - Abstract
Despite the initial successes of immunotherapy, there is an urgent clinical need for molecular assays that identify patients more likely to respond. Here, we report that ultrasensitive measures of circulating tumor DNA (ctDNA) and T-cell expansion can be used to assess responses to immune checkpoint blockade in metastatic lung cancer patients (N = 24). Patients with clinical response to therapy had a complete reduction in ctDNA levels after initiation of therapy, whereas nonresponders had no significant changes or an increase in ctDNA levels. Patients with initial response followed by acquired resistance to therapy had an initial drop followed by recrudescence in ctDNA levels. Patients without a molecular response had shorter progression-free and overall survival compared with molecular responders [5.2 vs. 14.5 and 8.4 vs. 18.7 months; HR 5.36; 95% confidence interval (CI), 1.57–18.35; P = 0.007 and HR 6.91; 95% CI, 1.37–34.97; P = 0.02, respectively], which was detected on average 8.7 weeks earlier and was more predictive of clinical benefit than CT imaging. Expansion of T cells, measured through increases of T-cell receptor productive frequencies, mirrored ctDNA reduction in response to therapy. We validated this approach in an independent cohort of patients with early-stage non–small cell lung cancer (N = 14), where the therapeutic effect was measured by pathologic assessment of residual tumor after anti-PD1 therapy. Consistent with our initial findings, early ctDNA dynamics predicted pathologic response to immune checkpoint blockade. These analyses provide an approach for rapid determination of therapeutic outcomes for patients treated with immune checkpoint inhibitors and have important implications for the development of personalized immune targeted strategies. Significance: Rapid and sensitive detection of circulating tumor DNA dynamic changes and T-cell expansion can be used to guide immune targeted therapy for patients with lung cancer. See related commentary by Zou and Meyerson, p. 1038
- Published
- 2019
28. Persistent mutant oncogene specific T cells in two patients benefitting from anti-PD-1
- Author
-
Qingfeng Zhu, Haidan Guo, Franco Verde, Hao Wang, Patrick M. Forde, Dung T. Le, Sune Justesen, Teniola Oke, Hongni Fan, Prerna Suri, Leslie Cope, Ludmila Danilova, Janis M. Taube, Anas H. Awan, Franck Housseau, Jennifer N. Durham, Hok Yee Chan, Nickolas Papadopoulos, Elizabeth D. Thompson, Bjarne Bartlett, Robert A. Anders, Nicholas Siegel, Victor E. Velculescu, Kellie N. Smith, John-William Sidhom, Laveet K. Aulakh, Kristen A. Marrone, Valsamo Anagnostou, Cynthia L. Sears, Joanne Riemer, Drew M. Pardoll, Luis A. Diaz, Brandon Luber, Ada J. Tam, Bert Vogelstein, Tricia R. Cottrell, Nicolas J. Llosa, Kenneth W. Kinzler, Julie R. Brahmer, William H. Sharfman, and Jarushka Naidoo
- Subjects
0301 basic medicine ,Cancer Research ,Lung Neoplasms ,Colorectal cancer ,T cell ,T-Lymphocytes ,Immunology ,Cell ,T cells ,Case Report ,Disease ,lcsh:RC254-282 ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Predictive biomarkers ,Checkpoint blockade ,Carcinoma, Non-Small-Cell Lung ,medicine ,Immunology and Allergy ,Humans ,Oncogene ,Aged ,Pharmacology ,Neoantigens ,business.industry ,Correction ,Oncogenes ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Primary tumor ,3. Good health ,Blockade ,030104 developmental biology ,medicine.anatomical_structure ,Nivolumab ,Treatment Outcome ,Oncology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Molecular Medicine ,DNA mismatch repair ,Female ,business ,Colorectal Neoplasms - Abstract
Background Several predictive biomarkers are currently approved or are under investigation for the selection of patients for checkpoint blockade. Tumor PD-L1 expression is used for stratification of non-small cell lung (NSCLC) patients, with tumor mutational burden (TMB) also being explored with promising results, and mismatch-repair deficiency is approved for tumor site-agnostic disease. While tumors with high PD-L1 expression, high TMB, or mismatch repair deficiency respond well to checkpoint blockade, tumors with lower PD-L1 expression, lower mutational burdens, or mismatch repair proficiency respond much less frequently. Case presentation We studied two patients with unexpected responses to checkpoint blockade monotherapy: a patient with PD-L1-negative and low mutational burden NSCLC and one with mismatch repair proficient colorectal cancer (CRC), both of whom lack the biomarkers associated with response to checkpoint blockade, yet achieved durable clinical benefit. Both maintained T-cell responses in peripheral blood to oncogenic driver mutations – BRAF-N581I in the NSCLC and AKT1-E17K in the CRC – years after treatment initiation. Mutation-specific T cells were also found in the primary tumor and underwent dynamic perturbations in the periphery upon treatment. Conclusions These findings suggest that T cell responses to oncogenic driver mutations may be more prevalent than previously appreciated and could be harnessed in immunotherapeutic treatment, particularly for patients who lack the traditional biomarkers associated with response. Comprehensive studies are warranted to further delineate additional predictive biomarkers and populations of patients who may benefit from checkpoint blockade. Electronic supplementary material The online version of this article (10.1186/s40425-018-0492-x) contains supplementary material, which is available to authorized users.
- Published
- 2019
29. Abstract 463: Highly accurate machine learning assessment of immune-related pathologic response criteria (irPRC) scoring in patients with non-small cell lung carcinoma (NSCLC) treated with neoadjuvant anti-PD-1-based therapies
- Author
-
Julie E. Stein, Vinay Pulim, Tricia R. Cottrell, Patrick M. Forde, and Janis M. Taube
- Subjects
Cancer Research ,Oncology - Abstract
Pathological complete response (no residual viable tumor, RVT) and/or major pathologic response (≤10% RVT) are now primary or secondary endpoints for a large proportion of clinical trials studying neoadjuvant immunotherapeutic regimens. We previously developed a scoring system for assessing pathologic response after immunotherapy, termed irPRC (Cottrell et al. Ann Oncol 2018). By these criteria, %RVT is assessed by dividing RVT by the sum of the surface area on the slide composed of RVT + necrosis + regression bed– the latter feature is where the tumor used to be and is characterized by fibroinflammatory stroma that is distinct from tumoral stroma. We have previously reported high inter-observer reproducibility for pathologic response assessment following immunotherapy. However, these assessments involve performing evaluations that are currently outside the scope of routine surgical pathology training and may be time-consuming. To date, these assessments have primarily been performed by academic pathologists who have seen the largest number of these cases as a part of clinical trials. A machine learning (ML)-powered assessment of irPRC would allow for faster, standardized evaluation and expanded access to patients treated outside of large academic centers. We trained a supervised convolutional neural network to assess pathologic response using irPRC on n=92 H&E-stained slides from patients with advanced, resectable NSCLC treated with neoadjuvant anti-PD-1 +/- anti-CTLA-4 at a single institution. The ML algorithm was trained based on ground-truth manual annotations by pathologists on whole slide digital scans and tested using leave-one-out cross validation. Each of ~830,000 image tiles was classified into one of four classes: tumor, necrosis, immune-mediated regression, or background lung tissue. Receiver operating curves showed that the algorithm exhibited high accuracy for predicting the various tissue classes with an area under the curve of 0.95, 0.96, 0.90, and 0.90 for the four classes, respectively. %RVT was calculated by dividing the surface area of RVT by total tumor bed surface area (RVT + necrosis + regression). There was a strong positive correlation between the machine assessed RVT and the human assessed RVT at both the slide level and case level (aggregate %RVT based on surface area from all slides for a given patient), Pearson’s r=0.95 and r=0.99, respectively. Here, we demonstrate that a ML algorithm performs as well as an experienced pathologist assessment in scoring pathologic response. These findings will need to be validated in larger studies. Additionally, the association of pathologic response with longer term patient outcomes will be evaluated as survival data matures to determine whether pathologic response is a robust surrogate of survival. Citation Format: Julie E. Stein, Vinay Pulim, Tricia R. Cottrell, Patrick M. Forde, Janis M. Taube. Highly accurate machine learning assessment of immune-related pathologic response criteria (irPRC) scoring in patients with non-small cell lung carcinoma (NSCLC) treated with neoadjuvant anti-PD-1-based therapies [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 463.
- Published
- 2022
30. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers
- Author
-
Alyza M. Skaist, Patrick M. Forde, Rulin Wang, Jiajia Zhang, Malcolm V. Brock, Zineb Belcaid, Haidan Guo, Victor E. Velculescu, Joshua E. Reuss, Matthew J. Bott, Matthew D. Hellmann, Sarah J. Wheelan, Boyang Zhang, Kristen A. Marrone, Peter B. Illei, Ajay Vaghasia, Richard L. Blosser, Sune Justesen, Bert Vogelstein, Jamie E. Chaft, Sampriti Thapa, Errol L. Bush, Srinivasan Yegnasubramanian, Janis M. Taube, Julie R. Brahmer, Tricia R. Cottrell, Luis Aparicio, Jennifer Meyers, Kornel E. Schuebel, Hok Yee Chan, Taha Merghoub, Zhicheng Ji, Wenpin Hou, David R. Jones, Margueritta El Asmar, Bernard J. Park, Jarushka Naidoo, Kellie N. Smith, Jinny Ha, Kenneth W. Kinzler, Ada Tam, Dipika Singh, Brian J. Mog, Justina X. Caushi, Shibin Zhou, Valsamo Anagnostou, Hongkai Ji, Emily Han-Chung Hsiue, Mara Lanis, Poromendro Burman, Begum Choudhury, Drew M. Pardoll, Arbor G. Dykema, Anuj Gupta, and Laurene S. Cheung
- Subjects
0301 basic medicine ,Lung Neoplasms ,T-Lymphocytes ,T cell ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Biology ,CD8-Positive T-Lymphocytes ,Article ,03 medical and health sciences ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Antigen ,Single-cell analysis ,Antigens, Neoplasm ,Neoplasms ,Carcinoma, Non-Small-Cell Lung ,medicine ,Tumor Microenvironment ,Cytotoxic T cell ,Humans ,T-cell receptor ,RNA-Seq ,CD8-positive T cells ,Author Correction ,Immune Checkpoint Inhibitors ,Cells, Cultured ,Tumor microenvironment ,Multidisciplinary ,Receptors, Interleukin-7 ,Immunotherapy ,Cellular immunity ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cancer research ,Tumour immunology ,Single-Cell Analysis ,Transcriptome ,Immunologic Memory ,CD8 - Abstract
PD-1 blockade unleashes CD8 T cells1, including those specific for mutation-associated neoantigens (MANA), but factors in the tumour microenvironment can inhibit these T cell responses. Single-cell transcriptomics have revealed global T cell dysfunction programs in tumour-infiltrating lymphocytes (TIL). However, the majority of TIL do not recognize tumour antigens2, and little is known about transcriptional programs of MANA-specific TIL. Here, we identify MANA-specific T cell clones using the MANA functional expansion of specific T cells assay3 in neoadjuvant anti-PD-1-treated non-small cell lung cancers (NSCLC). We use their T cell receptors as a ‘barcode’ to track and analyse their transcriptional programs in the tumour microenvironment using coupled single-cell RNA sequencing and T cell receptor sequencing. We find both MANA- and virus-specific clones in TIL, regardless of response, and MANA-, influenza- and Epstein–Barr virus-specific TIL each have unique transcriptional programs. Despite exposure to cognate antigen, MANA-specific TIL express an incompletely activated cytolytic program. MANA-specific CD8 T cells have hallmark transcriptional programs of tissue-resident memory (TRM) cells, but low levels of interleukin-7 receptor (IL-7R) and are functionally less responsive to interleukin-7 (IL-7) compared with influenza-specific TRM cells. Compared with those from responding tumours, MANA-specific clones from non-responding tumours express T cell receptors with markedly lower ligand-dependent signalling, are largely confined to HOBIThigh TRM subsets, and coordinately upregulate checkpoints, killer inhibitory receptors and inhibitors of T cell activation. These findings provide important insights for overcoming resistance to PD-1 blockade., Single-cell RNA sequencing and T cell receptor sequencing are combined to identify transcriptional programs specific to mutation-associated neoantigen-specific T cells in non-small cell lung cancers treated with anti-PD-1, providing insights into resistance to PD-1 blockade.
- Published
- 2021
31. Neoadjuvant nivolumab plus ipilimumab in resectable non-small cell lung cancer
- Author
-
Daphne Wang, Taha Merghoub, Jarushka Naidoo, Jinny Ha, David R. Jones, Marianna Zahurak, Joseph C. Murray, Ben Levy, Kellie N. Smith, Stephen R. Broderick, Drew M. Pardoll, Suzanne L. Topalian, Jamie E. Chaft, Richard J. Battafarano, Valsamo Anagnostou, Errol L. Bush, Janis M. Taube, Victor E. Velculescu, Mara Lanis, Peter B. Illei, Matthew D. Hellmann, Malcolm V. Brock, Gary L. Rosner, Hok Yee Chan, James R. White, Franco Verde, Joshua E. Reuss, Caroline G. McCarthy, Patrick M. Forde, Tricia R. Cottrell, Stephen C. Yang, and Julie R. Brahmer
- Subjects
0301 basic medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,medicine.medical_treatment ,Immunology ,Short Report ,Ipilimumab ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,Immunology and Allergy ,Medicine ,tumor microenvironment ,Humans ,Lung cancer ,Adverse effect ,RC254-282 ,Aged ,Pharmacology ,clinical trials as topic ,business.industry ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunotherapy ,Middle Aged ,medicine.disease ,Neoadjuvant Therapy ,030104 developmental biology ,Nivolumab ,Tumor progression ,030220 oncology & carcinogenesis ,tumor biomarkers ,Molecular Medicine ,Female ,KRAS ,immunotherapy ,business ,medicine.drug - Abstract
BackgroundWe conducted the first trial of neoadjuvant PD-1 blockade in resectable non-small cell lung cancer (NSCLC), finding nivolumab monotherapy to be safe and feasible with an encouraging rate of pathologic response. Building on these results, and promising data for nivolumab plus ipilimumab (anti-CTLA-4) in advanced NSCLC, we expanded our study to include an arm investigating neoadjuvant nivolumab plus ipilimumab.MethodsPatients with resectable stage IB (≥4 cm)–IIIA (American Joint Committee on Cancer Tumor Node Metastases seventh edition), histologically confirmed, treatment-naïve NSCLC received nivolumab 3 mg/kg intravenously plus ipilimumab 1 mg/kg intravenously 6 weeks prior to planned resection. Nivolumab 3 mg/kg was given again approximately 4 and 2 weeks preoperatively. Primary endpoints were safety and feasibility with a planned enrollment of 15 patients. Pathologic response was a key secondary endpoint.ResultsWhile the treatment regimen was feasible per protocol, due to toxicity, the study arm was terminated early by investigator consensus after 9 of 15 patients were enrolled. All patients received every scheduled dose of therapy and were fit for planned surgery; however, 6 of 9 (67%) experienced treatment-related adverse events (TRAEs) and 3 (33%) experienced grade ≥3 TRAEs. Three of 9 patients (33%) had biopsy-confirmed tumor progression precluding definitive surgery. Of the 6 patients who underwent resection, 3 are alive and disease-free, 2 experienced recurrence and are actively receiving systemic treatment, and one died postoperatively due to acute respiratory distress syndrome. Two patients who underwent resection had tumor pathologic complete responses (pCRs) and continue to remain disease-free over 24 months since surgery. Pathologic response correlated with pre-treatment tumor PD-L1 expression, but not tumor mutation burden. Tumor KRAS/STK11 co-mutations were identified in 5 of 9 patients (59%), of whom two with disease progression precluding surgery had tumor KRAS/STK11/KEAP1 co-mutations.ConclusionsThough treatment was feasible, due to toxicity the study arm was terminated early by investigator consensus. In light of this, and while the long-term disease-free status of patients who achieved pCR is encouraging, further investigation of neoadjuvant nivolumab plus ipilimumab in patients with resectable NSCLC requires the identification of predictive biomarkers that enrich for response.
- Published
- 2020
32. Chronic immune checkpoint inhibitor pneumonitis
- Author
-
Lonny Yarmus, Patrick M. Forde, Janis M. Taube, Daphne Wang, Joanne Riemer, Sonye K. Danoff, Karthik Suresh, Peter B. Illei, K. Ranh Voong, Evan J. Lipson, Jarushka Naidoo, Hans J. Lee, Tricia R. Cottrell, Cheng Ting Lin, David Feller-Kopman, Franco R. D'Alessio, and Julie R. Brahmer
- Subjects
Male ,Cancer Research ,medicine.medical_specialty ,Lymphocytosis ,medicine.medical_treatment ,Immunology ,Lung biopsy ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Prednisone ,Internal medicine ,medicine ,Humans ,Immunology and Allergy ,Adverse effect ,Immune Checkpoint Inhibitors ,RC254-282 ,Aged ,Retrospective Studies ,Pneumonitis ,Pharmacology ,Lung ,medicine.diagnostic_test ,business.industry ,Correction ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,Pneumonia ,Middle Aged ,medicine.disease ,Bronchoalveolar lavage ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Molecular Medicine ,Female ,medicine.symptom ,business ,hormones, hormone substitutes, and hormone antagonists ,030215 immunology ,medicine.drug - Abstract
BackgroundPneumonitis from immune checkpoint inhibitors (ICI) is a potentially fatal immune-related adverse event (irAE) from antiprogrammed death 1/programmed death ligand 1 immunotherapy. Most cases of ICI pneumonitis improve or resolve with 4–6 weeks of corticosteroid therapy. Herein, we report the incidence, clinicopathological features and management of patients with non-small cell lung cancer (NSCLC) and melanoma who developed chronic ICI pneumonitis that warrants ≥12 weeks of immunosuppression.MethodsPatients with ICI pneumonitis were identified from institutional databases of ICI-treated patients with advanced melanoma and NSCLC between January 2011 and July 2018. ICI pneumonitis was defined as clinical/radiographic evidence of lung inflammation without alternative diagnoses, adjudicated by a multidisciplinary team. Chronic ICI pneumonitis was defined as pneumonitis that persists or worsens with steroid tapering, and necessitates ≥12 weeks of immunosuppression, after ICI discontinuation. Serial chest CT was used to assess radiological features, and tumor response by Response EvaluationCriteria for Solid Tumors V.1.1. Bronchoalveolar lavage fluid (BALF) samples were assessed by cell differential. Lung biopsy samples were evaluated by H&E staining and multiplex immunofluorescence (mIF), where available.ResultsAmong 299 patients, 44 developed ICI pneumonitis (NSCLC: 5/205; melanoma: 1/94), and of these, 6 experienced chronic ICI pneumonitis. The overall incidence of chronic ICI pneumonitis was thus 2%. Of those who developed chronic ICI pneumonitis: the majority had NSCLC (5/6), all sustained disease control from ICIs, and none had other concurrent irAEs. Timing of chronic ICI pneumonitis development was variable (range: 0–50 months), and occurred at a median of 12 months post ICI start. Recrudescence of ICI pneumonitis occurred at a median of 6 weeks after initial steroid start (range: 3–12 weeks), with all patients requiring steroid reintroduction when tapered to ≤10 mg prednisone/equivalent. The median total duration of steroids was 37 weeks (range: 16–43+weeks). Re-emergence of radiographic ICI pneumonitis occurred in the same locations on chest CT, in most cases (5/6). All patients who developed chronic ICI pneumonitis had BALF lymphocytosis on cell differential and organising pneumonia on lung biopsy at initial ICI pneumonitis presentation, with persistent BALF lymphocytosis and brisk CD8+ infiltration on mIF at pneumonitis re-emergence during steroid taper.ConclusionsA subset of patients who develop pneumonitis from ICIs will develop chronic ICI pneumonitis, that warrants long-term immunosuppression of ≥12 weeks, and has distinct clinicopathological features.
- Published
- 2020
33. Evaluation of Peritumoral Fibrosis in Metastatic Colorectal Adenocarcinoma to the Liver Using Digital Image Analysis
- Author
-
Qingfeng Zhu, Sepideh Besharati, Robert A. Anders, Tricia R. Cottrell, and Kevin M. Waters
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Pathology, Surgical ,Adenocarcinoma ,Chronic liver disease ,Metastatic carcinoma ,03 medical and health sciences ,0302 clinical medicine ,Methyl Green ,Fibrosis ,Interquartile range ,Trichrome ,Image Processing, Computer-Assisted ,medicine ,Humans ,Colorectal adenocarcinoma ,Coloring Agents ,Aged ,business.industry ,Liver Neoplasms ,Original Articles ,General Medicine ,Middle Aged ,medicine.disease ,Immunohistochemistry ,Liver ,030220 oncology & carcinogenesis ,Digital image analysis ,Eosine Yellowish-(YS) ,Female ,Colorectal Neoplasms ,business ,Azo Compounds ,Software ,030215 immunology - Abstract
Objectives It is challenging to separate peritumoral fibrosis from fibrosis due to chronic liver disease in mass-directed liver biopsies. We evaluated the distance that peritumoral fibrosis extends from metastatic colorectal adenocarcinoma in liver. Methods Peritumoral and distant uninvolved liver trichrome stains from 25 cases were analyzed using digital image analysis. Fibrosis was quantitated at concentric intervals from each tumor and in uninvolved liver. Results There was a 3.9 fold (range 0.9-18.6) median increase in fibrosis in the first 0.5 mm of peritumoral liver compared to distant liver. Fibrosis levels returned to baseline at median 2.5 mm (interquartile range 1.5-5.0 mm) from tumor. Conclusions Fibrosis is markedly increased in peritumoral liver. Fibrosis levels returned to baseline by 5 mm from tumor in approximately 75% of cases. Pathologists should be cautious of fibrosis in mass-directed liver biopsies without at least 5 mm of liver tissue distal to the mass.
- Published
- 2018
34. Multidimensional, quantitative assessment of PD-1/PD-L1 expression in patients with Merkel cell carcinoma and association with response to pembrolizumab
- Author
-
Benjamin Green, Aleksandra Ogurtsova, Paul Nghiem, Jennifer H. Yearley, Elizabeth L. Engle, Nirasha Ramchurren, Candice D. Church, Abha Soni, Genevieve J. Kaunitz, Steve Fling, Joel C. Sunshine, Julie E. Stein, Ludmila Danilova, Janis M. Taube, Farah Succaria, Tricia R. Cottrell, Natalie J. Miller, Peter Nguyen, Haiying Xu, Nicolas A. Giraldo, Lisa Lundgren, Robert A. Anders, Mac Cheever, Jonathan D. Cuda, Suzanne L. Topalian, Evan J. Lipson, and Sneha Berry
- Subjects
Male ,0301 basic medicine ,PD-L1 ,Cancer Research ,Cell type ,Multispectral immunofluorescence ,Programmed Cell Death 1 Receptor ,Immunology ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,Immunofluorescence ,lcsh:RC254-282 ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,0302 clinical medicine ,PD-1 ,medicine ,Humans ,Immunology and Allergy ,Pharmacology ,CD20 ,Merkel cell ,biology ,medicine.diagnostic_test ,Merkel cell carcinoma ,business.industry ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Carcinoma, Merkel Cell ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Molecular Medicine ,Female ,business ,CD8 ,Research Article - Abstract
Background We recently reported a 56% objective response rate in patients with advanced Merkel cell carcinoma (MCC) receiving pembrolizumab. However, a biomarker predicting clinical response was not identified. Methods Pretreatment FFPE tumor specimens (n = 26) were stained for CD8, PD-L1, and PD-1 by immunohistochemistry/immunofluorescence (IHC/IF), and the density and distribution of positive cells was quantified to determine the associations with anti-PD-1 response. Multiplex IF was used to test a separate cohort of MCC archival specimens (n = 16), to identify cell types expressing PD-1. Results Tumors from patients who responded to anti-PD-1 showed higher densities of PD-1+ and PD-L1+ cells when compared to non-responders (median cells/mm2, 70.7 vs. 6.7, p = 0.03; and 855.4 vs. 245.0, p = 0.02, respectively). There was no significant association of CD8+ cell density with clinical response. Quantification of PD-1+ cells located within 20 μm of a PD-L1+ cell showed that PD-1/PD-L1 proximity was associated with clinical response (p = 0.03), but CD8/PD-L1 proximity was not. CD4+ and CD8+ cells in the TME expressed similar amounts of PD-1. Conclusions While the binomial presence or absence of PD-L1 expression in the TME was not sufficient to predict response to anti-PD-1 in patients with MCC, we show that quantitative assessments of PD-1+ and PD-L1+ cell densities as well as the geographic interactions between these two cell populations correlate with clinical response. Cell types expressing PD-1 in the TME include CD8+ T-cells, CD4+ T-cells, Tregs, and CD20+ B-cells, supporting the notion that multiple cell types may potentiate tumor regression following PD-1 blockade. Electronic supplementary material The online version of this article (10.1186/s40425-018-0404-0) contains supplementary material, which is available to authorized users.
- Published
- 2018
35. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer
- Author
-
Franco Verde, Kristen A. Marrone, Edward Gabrielson, Taha Merghoub, Gary L. Rosner, Richard J. Battafarano, Marianna Zahurak, Justina X. Caushi, Patrick M. Forde, Jamie E. Chaft, Natasha Rekhtman, Moises J. Velez, Robert B. Scharpf, Jarushka Naidoo, David R. Jones, Tricia R. Cottrell, Jiajia Zhang, Janis M. Taube, John-William Sidhom, Zachary T. Olah, Julie R. Brahmer, James R. White, Hok Yee Chan, Kellie N. Smith, Stephen C. Yang, Victor E. Velculescu, Valerie W. Rusch, Stephen R. Broderick, Drew M. Pardoll, Valsamo Anagnostou, Jedd D. Wolchok, Hao Wang, Haidan Guo, Matthew D. Hellmann, and Suzanne L. Topalian
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,General Medicine ,medicine.disease ,Article ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Major Pathologic Response ,030220 oncology & carcinogenesis ,Internal medicine ,Biopsy ,Carcinoma ,medicine ,Pd 1 blockade ,Nivolumab ,business ,Lung cancer ,Neoadjuvant therapy - Abstract
Background Antibodies that block programmed death 1 (PD-1) protein improve survival in patients with advanced non–small-cell lung cancer (NSCLC) but have not been tested in resectable NSCLC, a condition in which little progress has been made during the past decade. Methods In this pilot study, we administered two preoperative doses of PD-1 inhibitor nivolumab in adults with untreated, surgically resectable early (stage I, II, or IIIA) NSCLC. Nivolumab (at a dose of 3 mg per kilogram of body weight) was administered intravenously every 2 weeks, with surgery planned approximately 4 weeks after the first dose. The primary end points of the study were safety and feasibility. We also evaluated the tumor pathological response, expression of programmed death ligand 1 (PD-L1), mutational burden, and mutation-associated, neoantigen-specific T-cell responses. Results Neoadjuvant nivolumab had an acceptable side-effect profile and was not associated with delays in surgery. Of the 21 tumors that were removed...
- Published
- 2018
36. Implications of the tumor immune microenvironment for staging and therapeutics
- Author
-
Robert A. Anders, Nicolas A. Giraldo, Janis M. Taube, Sanjay S. Patel, Alexander S. Baras, Jérôme Galon, Scott J. Rodig, Lynette M. Sholl, Ashley Cimino-Mathews, David L. Rimm, and Tricia R. Cottrell
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Immune microenvironment ,Article ,Pathology and Forensic Medicine ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Neoplasms ,Cell density ,Biomarkers, Tumor ,Tumor Microenvironment ,medicine ,Humans ,Cytotoxic T cell ,Stage (cooking) ,Neoplasm Staging ,Tumor microenvironment ,biology ,business.industry ,Prognosis ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Antibody ,business ,CD8 - Abstract
Characterizing the tumor immune microenvironment enables the identification of new prognostic and predictive biomarkers, the development of novel therapeutic targets and strategies, and the possibility to guide first-line treatment algorithms. Although the driving elements within the tumor microenvironment of individual primary organ sites differ, many of the salient features remain the same. The presence of a robust antitumor milieu characterized by an abundance of CD8+ cytotoxic T-cells, Th1 helper cells, and associated cytokines often indicates a degree of tumor containment by the immune system and can even lead to tumor elimination. Some of these features have been combined into an ‘Immunoscore’, which has been shown to complement the prognostic ability of the current TNM staging for early stage colorectal carcinomas. Features of the immune microenvironment are also potential therapeutic targets, and immune checkpoint inhibitors targeting the PD-1/ PD-L1 axis are especially promising. FDA-approved indications for anti-PD-1/PD-L1 are rapidly expanding across numerous tumor types and, in certain cases, are accompanied by companion or complimentary PD-L1 immunohistochemical diagnostics. Pathologists have direct visual access to tumor tissue and in-depth knowledge of the histological variations between and within tumor types and thus are poised to drive forward our understanding of the tumor microenvironment. This review summarizes the key components of the tumor microenvironment, presents an overview of and the challenges with PD-L1 antibodies and assays, and addresses newer candidate biomarkers, such as CD8+ cell density and mutational load. Characteristics of the local immune contexture and current pathology-related practices for specific tumor types are also addressed. In the future, characterization of the host antitumor immune response using multiplexed and multimodality biomarkers may help predict which patients will respond to immune-based therapies.
- Published
- 2018
37. PD-L1 and Emerging Biomarkers in Immune Checkpoint Blockade Therapy
- Author
-
Janis M. Taube and Tricia R. Cottrell
- Subjects
0301 basic medicine ,Cancer Research ,Programmed Cell Death 1 Receptor ,medicine.disease_cause ,Stain ,Article ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Neoplasms ,PD-L1 ,Biomarkers, Tumor ,Tumor Microenvironment ,Humans ,Cytotoxic T cell ,Medicine ,Tumor microenvironment ,Mutation ,biology ,business.industry ,Immune checkpoint ,Tumor Burden ,Blockade ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Immunohistochemistry ,business - Abstract
PD-L1 checkpoint blockade is revolutionizing cancer therapy, and biomarkers capable of predicting which patients are most likely to respond are highly desired. The detection of PD-L1 protein expression by immunohistochemistry can enrich for response to anti-PD-(L)1 blockade in a variety of tumor types, but is not absolute. Limitations of current commercial PD-L1 immunohistochemical (IHC) assays and improvements anticipated in next-generation PD-L1 testing are reviewed. Assessment of tumor-infiltrating lymphocytes in conjunction with PD-L1 testing could improve specificity by distinguishing adaptive (interferon γ driven and cytotoxic T-lymphocyte associated) from constitutive (non-immune mediated) expression. The presence of a high tumor mutational burden also enriches for response to therapy, and early data indicate that this may provide additive predictive value beyond PD-L1 IHC alone. As candidate biomarkers continue to emerge, the pathologist's assessment of the tumor microenvironment on hematoxylin-eosin stain combined with PD-L1 IHC remains a rapid and robust way to evaluate the tumor-immune dynamic.
- Published
- 2018
38. Author Correction: Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers
- Author
-
Ajay Vaghasia, Anuj Gupta, Jamie E. Chaft, Hok Yee Chan, Laurene S. Cheung, Malcolm V. Brock, Sarah J. Wheelan, Emily Han-Chung Hsiue, Taha Merghoub, Jiajia Zhang, Wenpin Hou, David R. Jones, Alyza M. Skaist, Srinivasan Yegnasubramanian, Sampriti Thapa, Luis Aparicio, Valsamo Anagnostou, Margueritta El Asmar, Kellie N. Smith, Dipika Singh, Mara Lanis, Zineb Belcaid, Sune Justesen, Haidan Guo, Patrick M. Forde, Kornel E. Schuebel, Matthew D. Hellmann, Begum Choudhury, Matthew J. Bott, Kenneth W. Kinzler, Bert Vogelstein, Jennifer Meyers, Boyang Zhang, Bernard J. Park, Rulin Wang, Janis M. Taube, Tricia R. Cottrell, Kristen A. Marrone, Julie R. Brahmer, Poromendro Burman, Zhicheng Ji, Brian J. Mog, Jarushka Naidoo, Jinny Ha, Drew M. Pardoll, Richard L. Blosser, Arbor G. Dykema, Justina X. Caushi, Hongkai Ji, Errol L. Bush, Ada Tam, Shibin Zhou, Victor E. Velculescu, Joshua E. Reuss, and Peter B. Illei
- Subjects
Cellular immunity ,Multidisciplinary ,Lung ,medicine.anatomical_structure ,business.industry ,medicine.medical_treatment ,Anti pd 1 ,T-cell receptor ,medicine ,Cancer research ,Immunotherapy ,business ,Tumor immunology - Published
- 2021
39. Melanoma subtypes demonstrate distinct PD-L1 expression profiles
- Author
-
Robert A. Anders, Meg R. Gerstenblith, Genevieve J. Kaunitz, Evan J. Lipson, Charles G. Eberhart, Sneha Berry, Valliammai Muthappan, Cheryl L. Thompson, Janis M. Taube, Jessica Esandrio, James T. Handa, Alexander H. Fischer, Stefan Kraft, Aleksandra Ogurtsova, Haiying Xu, Kord Honda, Jonathan D. Cuda, Mohammed Lilo, and Tricia R. Cottrell
- Subjects
Uveal Neoplasms ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,Skin Neoplasms ,Lymphocyte ,Uveal Neoplasm ,Lentigo maligna ,Biology ,Cell morphology ,B7-H1 Antigen ,Article ,Pathology and Forensic Medicine ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Melanoma ,Molecular Biology ,Skin ,Gene Expression Profiling ,Cell Biology ,medicine.disease ,Gene expression profiling ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cutaneous melanoma ,Immunohistochemistry - Abstract
PD-L1 expression in the tumor immune microenvironment is recognized as both a prognostic and predictive biomarker in patients with cutaneous melanoma, a finding closely related to its adaptive (IFN-γ-mediated) mechanism of expression. Approximately 35% of cutaneous melanomas express PD-L1, however, the expression patterns, levels, and prevalence in rarer melanoma subtypes are not well described. We performed immunohistochemistry for PD-L1 and CD8 on 200 formalin-fixed paraffin-embedded specimens from patients with acral (n = 16), mucosal (n = 36), uveal (n = 103), and chronic sun-damaged (CSD) (n = 45) melanomas (24 lentigo maligna, 13 ‘mixed’ desmoplastic, and 8 ‘pure’ desmoplastic melanomas). CD8+ tumor-infiltrating lymphocyte (TIL) densities were characterized as mild, moderate, or severe, and their geographic association with PD-L1 expression was evaluated. Discrete lymphoid aggregates, the presence of a spindle cell morphology, and the relationship of these features with PD-L1 expression were assessed. PD-L1 expression was observed in 31% of acral melanomas, 44% of mucosal melanomas, 10% of uveal melanomas, and 62% of CSD melanomas (P
- Published
- 2017
40. Transcriptional Mechanisms of Resistance to Anti–PD-1 Therapy
- Author
-
Leslie Cope, Nadeem Riaz, Christine A. Iacobuzio-Donahue, Janis M. Taube, Barry S. Taylor, Ralph H. Hruban, Suzanne L. Topalian, Jinshui Fan, Evan J. Lipson, Genevieve J. Kaunitz, Maria Libera Ascierto, Timothy A. Chan, Zachary A. Kohutek, David B. Solit, Alvin Makohon-Moore, Vladimir Makarov, Tricia R. Cottrell, Tracee L. McMiller, Alexander V. Favorov, Alan E. Berger, and Drew M. Pardoll
- Subjects
0301 basic medicine ,Cancer Research ,DNA Copy Number Variations ,Programmed Cell Death 1 Receptor ,Disease ,Drug resistance ,Biology ,Somatic evolution in cancer ,Article ,Clonal Evolution ,03 medical and health sciences ,Exome Sequencing ,Gene expression ,medicine ,Humans ,Neoplasm Metastasis ,Melanoma ,Exome sequencing ,Regulation of gene expression ,Cancer ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Treatment Outcome ,030104 developmental biology ,Oncology ,Drug Resistance, Neoplasm ,Immunology - Abstract
Purpose: To explore factors associated with response and resistance to anti–PD-1 therapy, we analyzed multiple disease sites at autopsy in a patient with widely metastatic melanoma who had a heterogeneous response. Materials and Methods: Twenty-six melanoma specimens (four premortem, 22 postmortem) were subjected to whole exome sequencing. Candidate immunologic markers and gene expression were assessed in 10 cutaneous metastases showing response or progression during therapy. Results: The melanoma was driven by biallelic inactivation of NF1. All lesions had highly concordant mutational profiles and copy number alterations, indicating linear clonal evolution. Expression of candidate immunologic markers was similar in responding and progressing lesions. However, progressing cutaneous metastases were associated with overexpression of genes associated with extracellular matrix and neutrophil function. Conclusions: Although mutational and immunologic differences have been proposed as the primary determinants of heterogeneous response/resistance to targeted therapies and immunotherapies, respectively, differential lesional gene expression profiles may also dictate anti–PD-1 outcomes. Clin Cancer Res; 23(12); 3168–80. ©2017 AACR. See related commentary by Wilmott et al., p. 2921
- Published
- 2017
41. Analysis of multispectral imaging with the AstroPath platform informs efficacy of PD-1 blockade
- Author
-
Ludmila Danilova, Haiying Xu, Alexander S. Baras, Qingfeng Zhu, Daphne Wang, Nicolas A. Giraldo, Danielle Signer, Janis M. Taube, Shlomit Jessel, Benjamin Green, Margaret Eminizer, Julie E. Stein, Charles Roberts, Jeffrey Roskes, Aleksandra Ogurtsova, Richard Wilton, Seyoun Park, Elizabeth L. Engle, Leslie Cope, Evan J. Lipson, Elizabeth M. Jaffee, Harriet M. Kluger, Robert A. Anders, David L. Rimm, Sneha Berry, Sigfredo Soto-Diaz, Angelo M. De Marzo, Inbal B. Sander, Pok Fai Wong, Drew M. Pardoll, Joel C. Sunshine, Jose Loyola, Tricia R. Cottrell, Peter Nguyen, Alexander S. Szalay, Joshua Doyle, and Suzanne L. Topalian
- Subjects
Adult ,Male ,Cell type ,medicine.medical_treatment ,CD8 Antigens ,Programmed Cell Death 1 Receptor ,Antigens, Differentiation, Myelomonocytic ,Fluorescent Antibody Technique ,Receptors, Cell Surface ,B7-H1 Antigen ,Antineoplastic Agents, Immunological ,Single-cell analysis ,Antigens, CD ,T-Lymphocyte Subsets ,Biomarkers, Tumor ,Tumor Microenvironment ,Medicine ,Humans ,Progression-free survival ,Melanoma ,Aged ,Aged, 80 and over ,Tumor microenvironment ,Multidisciplinary ,business.industry ,SOXE Transcription Factors ,Macrophages ,FOXP3 ,Forkhead Transcription Factors ,Immunotherapy ,Middle Aged ,medicine.disease ,Immune Checkpoint Proteins ,Prognosis ,Progression-Free Survival ,Blockade ,Treatment Outcome ,Cancer research ,Female ,Single-Cell Analysis ,business - Abstract
Astronomy accelerates tumor imaging Immunohistochemical stains for individual markers revolutionized diagnostic pathology decades ago but cannot capture enough information to accurately predict response to immunotherapy. Newer multiplex immunofluorescent technologies provide the potential to visualize the expression patterns of many functionally relevant molecules but present numerous challenges in accurate image analysis and data handling, particularly over large tumor areas. Drawing from the field of astronomy, in which petabytes of imaging data are routinely analyzed across a wide spectral range, Berry et al. developed a platform for multispectral imaging of whole-tumor sections with high-fidelity single-cell resolution. The resultant AstroPath platform was used to develop a multiplex immunofluorescent assay highly predictive of responses and outcomes for melanoma patients receiving immunotherapy. Science , aba2609, this issue p. eaba2609
- Published
- 2019
42. Enrichment of Scleroderma Vascular Disease-Associated Autoantigens in Endothelial Lineage Cells
- Author
-
Tricia R. Cottrell, Marc K. Halushka, Livia Casciola-Rosen, Antony Rosen, Tea Soon Park, Zsuzsanna H. McMahan, Elias T. Zambidis, Fredrick M. Wigley, Laura Gutierrez-Alamillo, Brendan Antiochos, and Raffaello Cimbro
- Subjects
030203 arthritis & rheumatology ,0301 basic medicine ,CD31 ,Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,Vascular disease ,Immunology ,Autoantibody ,Embryoid body ,Biology ,medicine.disease ,Scleroderma ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Rheumatology ,Biopsy ,medicine ,Immunology and Allergy ,Immunohistochemistry ,Progenitor cell - Abstract
Objective Scleroderma patients with autoantibodies to CENPs and/or interferon-inducible protein 16 (IFI-16) are at increased risk of severe vascular complications. This study was undertaken to determine whether these autoantigens are enriched in cells of the vasculature. Methods Successive stages of embryoid bodies (EBs) as well as vascular progenitors were used to evaluate the expression of scleroderma autoantigens IFI-16 and CENP by immunoblotting. CD31 was included to mark early blood vessels. IFI-16 and CD31 expression were defined in paraffin-embedded skin sections from scleroderma patients and from healthy controls. IFI-16 expression was determined by flow cytometric analysis in circulating endothelial cells (CECs) and circulating hematopoietic progenitor cells. Results Expression of CENP-A, IFI-16, and CD31 was enriched in EBs on days 10 and 12 of differentiation, and particularly in cultures enriched in vascular progenitors (IFI-16, CD31, and CENPs A and B). This pattern was distinct from that of comparator autoantigens. Immunohistochemical staining of paraffin-embedded skin sections showed enrichment of IFI-16 in CD31-positive vascular endothelial cells in biopsy specimens from scleroderma patients and normal controls. Flow cytometric analysis revealed IFI-16 expression in circulating hematopoietic progenitor cells but minimal expression in CECs. Conclusion Our findings indicate that expression of the scleroderma autoantigens IFI-16 and CENPs, which are associated with severe vascular disease, is increased in vascular progenitors and mature endothelial cells. High level, lineage-enriched expression of autoantigens may explain the striking association between clinical phenotypes and the immune targeting of specific autoantigens.
- Published
- 2016
43. Abstract 6584: The ‘AstroPath' platform for spatially resolved, single cell analysis of the tumor microenvironment (TME) using multispectral immunofluorescence (mIF)
- Author
-
Suzanne L. Topalian, Aleksandra Ogurtsova, Tricia R. Cottrell, Peter Nguyen, Haiying Xu, Alexander S. Szalay, Janis M. Taube, Angelo M. DeMarzo, Sneha Berry, Robert A. Anders, Drew M. Pardoll, Julie E. Stein, Benjamin Green, Daphne Wang, Elizabeth L. Engle, and Nicolas A. Giraldo
- Subjects
0301 basic medicine ,Cancer Research ,Tumor microenvironment ,medicine.diagnostic_test ,Computer science ,Spatially resolved ,Multispectral image ,Computational biology ,Gold standard (test) ,Immunofluorescence ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Single-cell analysis ,030220 oncology & carcinogenesis ,medicine ,Biomarker discovery ,Tumor marker - Abstract
Background: Multidimensional, spatially resolved analyses of immune and tumor cells within the TME of patients treated with checkpoint inhibitors will provide clinically translatable mechanistic insights and potentiate biomarker discovery. To achieve this goal, information from pathology specimens needs to be captured at a single cell level with high fidelity and in meaningfully sized cohorts. To date, efforts have been limited by inadequate tissue sampling and previously unrecognized errors in staining, imaging and data analysis. Here we describe the ‘AstroPath' platform, where strategies from the field of astronomy were adapted to study pathology specimens and generate large high quality mIF data. Methods: Potential error was identified and addressed at each stage of 6-plex (PD-1, PD-L1, FoxP3, CD163, CD8, tumor marker) mIF assay development. Whole slides from formalin-fixed paraffin embedded tissue specimens were stained with the optimized assay and imaged using a multispectral microscope (Vectra 3.0) with 20% overlap of high power fields (HPFs). The overlaps were used to quantify and correct optical lens distortion, HPF alignment, and illumination variation. Errors from cell segmentation algorithms, batch-to-batch staining variation, and HPF sampling were also addressed. The resultant mIF data were organized and analyzed using a large, relational database. Results: The optimized mIF assay captured equivalent signal compared to gold standard chromogenic immunohistochemistry and 2x more signal for PD-1, PD-L1 and FoxP3 compared to the manufacturer's recommended protocol. Errors and corrections for imaging included: pixel alignment error reduced from ~10 to Conclusion: Here we present an end-to-end pathology workflow with rigorous quality control for creating quantitative, spatially resolved mIF datasets using lessons derived from the field of astronomy. Such approaches will vastly improve standardization and scalability of mIF technologies, enabling cross-site comparisons and eventual clinical translation as biomarker discovery platforms or standard diagnostic tests. Citation Format: Sneha Berry, Nicolas Giraldo, Benjamin Green, Elizabeth Engle, Haiying Xu, Aleksandra Ogurtsova, Daphne Wang, Julie E. Stein, Peter Nguyen, Suzanne Topalian, Angelo DeMarzo, Drew M. Pardoll, Robert A. Anders, Tricia R. Cottrell, Alexander S. Szalay, Janis M. Taube. The ‘AstroPath' platform for spatially resolved, single cell analysis of the tumor microenvironment (TME) using multispectral immunofluorescence (mIF) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 6584.
- Published
- 2020
44. Trial in progress: Neoadjuvant immune checkpoint blockade in resectable malignant pleural mesothelioma
- Author
-
Tricia R. Cottrell, Stephen C. Yang, Julie E. Stein, Russell K. Hales, Marianna Zahurak, Peter B. Illei, Patrick M. Forde, Anne S. Tsao, Janis M. Taube, Richard J. Battafarano, Joseph Friedberg, Joshua E. Reuss, Erica C. Nakajima, Kellie N. Smith, Valsamo Anagnostou, Boris Sepesi, Joseph C. Murray, Gary L. Rosner, K. Ranh Voong, and Christian Rolfo
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Pleural mesothelioma ,business.industry ,Immune checkpoint ,Blockade ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,Medicine ,Stage (cooking) ,business ,030215 immunology - Abstract
TPS9078 Background: While the role of surgery in limited-stage (stage I-III) malignant pleural mesothelioma (MPM) is controversial, many centers have adopted an aggressive tri-modality approach incorporating (neo)adjuvant chemotherapy, surgical resection and radiotherapy. Despite this, most patients relapse and die from their disease. Immune checkpoint blockade (ICB) has shown promise in advanced MPM, but the mechanisms of response and resistance remain elusive. Improving the mechanistic understanding of ICB in MPM while concurrently optimizing the treatment strategy for limited-stage MPM are two urgent unmet needs. This multicenter multi-arm phase I/II study seeks to evaluate the safety and feasibility of neoadjuvant ICB in resectable MPM, incorporating novel genomic and immunologic analyses to deliver mechanistic insight into the biology of ICB in MPM. Methods: Patients with surgically resectable stage I-III treatment-naïve epithelioid or biphasic MPM receive neoadjuvant treatment with nivolumab every 2 weeks for 3 doses with or without 1 dose of ipilimumab (arm A: nivolumab monotherapy; arm B: nivolumab + ipilimumab). After macroscopic complete resection, patients receive optional investigator-choice adjuvant chemotherapy +/- radiation. Following this, patients will receive up to 1 year of adjuvant nivolumab. Feasibility and safety are co-primary endpoints of this study with feasibility defined by a delay in surgery of ≤24 days from the preplanned surgical date and safety defined by adverse events according to CTCAE v5.0. Bayesian-designed stopping rules have been implemented for feasibility and safety. Secondary endpoints include assessment of pathologic response and radiographic response using RECIST 1.1 for MPM. Correlative analyses will be performed on tissue specimens obtained pre- and post-ICB, as well as blood obtained pre, during, and post-ICB. Key correlates include multiplex immunofluorescence and longitudinal ctDNA assessment. Whole exome sequencing, T-cell receptor sequencing, and the MANAFEST functional neoantigen assay will be utilized to identify neoantigen-specific T-cell clonotypes and track these clonotypes temporally (during/post ICB) and spatially (across immune compartments). Single-cell RNA sequencing will be used to characterize the functionality of expanded T-cell clonotypes. Accrual to arm B will commence following complete accrual to arm A with a planned total enrollment of 30 patients. This study is open with 1 patient enrolled at the time of submission. Clinical trial information: NCT03918252.
- Published
- 2020
45. Pathologic features of response to neoadjuvant anti-PD-1 in resected non-small-cell lung carcinoma: a proposal for quantitative immune-related pathologic response criteria (irPRC)
- Author
-
Ed Gabrielson, Jennifer L. Sauter, Valsamo Anagnostou, Janis M. Taube, Robert A. Anders, Jedd D. Wolchok, Ashley Cimino-Mathews, Moises J. Velez, Kellie N. Smith, Suzanne L. Topalian, Amy S. Duffield, Patrick M. Forde, Frederic B. Askin, Ludmila Danilova, Richard J. Battafarano, Victor E. Velculescu, Drew M. Pardoll, Peter B. Illei, Matthew D. Hellmann, Jonathan D. Cuda, Noushin Niknafs, David R. Jones, Elizabeth D. Thompson, Julie E. Stein, Jamie E. Chaft, Natasha Rekhtman, James M. Isbell, Tricia R. Cottrell, Stephen C. Yang, and Julie R. Brahmer
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,Lung Neoplasms ,Neoplasm, Residual ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,H&E stain ,Breast Neoplasms ,Context (language use) ,03 medical and health sciences ,Pneumonectomy ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Major Pathologic Response ,Carcinoma, Non-Small-Cell Lung ,Antineoplastic Combined Chemotherapy Protocols ,Carcinoma ,Medicine ,Humans ,Lung ,Neoadjuvant therapy ,business.industry ,Reproducibility of Results ,Original Articles ,Hematology ,medicine.disease ,Prognosis ,Chemotherapy regimen ,Ipilimumab ,Neoadjuvant Therapy ,030104 developmental biology ,Nivolumab ,Treatment Outcome ,Editorial ,Oncology ,030220 oncology & carcinogenesis ,Feasibility Studies ,Immunotherapy ,Radiology ,business - Abstract
BACKGROUND: Neoadjuvant anti-PD-1 may improve outcomes for patients with resectable NSCLC and provides a critical window for examining pathologic features associated with response. Resections showing major pathologic response to neoadjuvant therapy, defined as ≤10% residual viable tumor (RVT), may predict improved long-term patient outcome. However, %RVT calculations were developed in the context of chemotherapy (%cRVT). An immune-related %RVT (%irRVT) has yet to be developed. PATIENTS AND METHODS: The first trial of neoadjuvant anti-PD-1 (nivolumab, NCT02259621) was just reported. We analyzed hematoxylin and eosin-stained slides from the post-treatment resection specimens of the 20 patients with non-small-cell lung carcinoma who underwent definitive surgery. Pretreatment tumor biopsies and preresection radiographic ‘tumor’ measurements were also assessed. RESULTS: We found that the regression bed (the area of immune-mediated tumor clearance) accounts for the previously noted discrepancy between CT imaging and pathologic assessment of residual tumor. The regression bed is characterized by (i) immune activation—dense tumor infiltrating lymphocytes with macrophages and tertiary lymphoid structures; (ii) massive tumor cell death—cholesterol clefts; and (iii) tissue repair—neovascularization and proliferative fibrosis (each feature enriched in major pathologic responders versus nonresponders, P
- Published
- 2018
46. MA11.10 Peripheral T Cell Repertoire Evolution in Resectable NSCLC Treated with Neoadjuvant PD-1 Blockade
- Author
-
Srinivasan Yegnasubramanian, Tricia R. Cottrell, Patrick M. Forde, Zhicheng Ji, Hok Yee Chan, Valsamo Anagnostou, Ed Gabrielson, Jarushka Naidoo, D. Pardoll, Janis M. Taube, Jamie E. Chaft, M. El Asmar, Jedd D. Wolchok, Hongkai Ji, Haidan Guo, Matthew D. Hellmann, Ni Zhao, Taha Merghoub, Kristen A. Marrone, Joshua E. Reuss, Justina X. Caushi, Julie R. Brahmer, Victor E. Velculescu, Jiajia Zhang, and Kellie N. Smith
- Subjects
Pulmonary and Respiratory Medicine ,T cell repertoire ,Oncology ,business.industry ,Cancer research ,Medicine ,Pd 1 blockade ,business ,Peripheral - Published
- 2019
47. Correction to: persistent mutant oncogene specific T cells in two patients benefitting from anti-PD-1
- Author
-
Qingfeng Zhu, Victor E. Velculescu, Bjarne Bartlett, Haidan Guo, Robert A. Anders, Drew M. Pardoll, Luis A. Diaz, William H. Sharfman, Brandon Luber, Prerna Suri, Nicholas Siegel, Valsamo Anagnostou, Sune Justesen, Patrick M. Forde, Hok Yee Chan, John-William Sidhom, Hao Wang, Franco Verde, Nickolas Papadopoulos, Dung T. Le, Teniola Oke, Bert Vogelstein, Elizabeth D. Thompson, Cynthia L. Sears, Hongni Fan, Tricia R. Cottrell, Kristen A. Marrone, Leslie Cope, Ada J. Tam, Ludmila Danilova, Anas H. Awan, Julie R. Brahmer, Jennifer N. Durham, Joanne Riemer, Jarushka Naidoo, Nicolas J. Llosa, Kenneth W. Kinzler, Janis M. Taube, Franck Housseau, Laveet K. Aulakh, and Kellie N. Smith
- Subjects
Pharmacology ,Cancer Research ,Oncogene ,business.industry ,Immunology ,Mutant ,Anti pd 1 ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,lcsh:RC254-282 ,Oncology ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,Medicine ,business - Published
- 2019
48. PD-L1 expression in inflammatory myofibroblastic tumors
- Author
-
Janis M. Taube, Aleksandra Ogurtsova, Anh T Duong, Haiying Xu, Tricia R. Cottrell, Christopher D. Gocke, and Deborah A. Belchis
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,medicine.drug_class ,T cell ,medicine.medical_treatment ,Myofibroma ,Clone (cell biology) ,Inflammation ,Soft Tissue Neoplasms ,Tyrosine-kinase inhibitor ,B7-H1 Antigen ,Article ,Pathology and Forensic Medicine ,Targeted therapy ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Biomarkers, Tumor ,Humans ,Child ,Aged ,business.industry ,Infant, Newborn ,Infant ,Middle Aged ,3. Good health ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Child, Preschool ,Immunohistochemistry ,Female ,medicine.symptom ,business ,CD8 - Abstract
Inflammatory myofibroblastic tumor is a rare mesenchymal tumor occurring at many anatomic sites, with a predilection for children and young adults. Often indolent, they can be locally aggressive and can metastasize, resulting in significant morbidity and mortality. Therapeutic options are often limited. The identification of underlying kinase mutations has allowed the use of targeted therapy in a subset of patients. Unfortunately, not all tumors harbor mutations and resistance to tyrosine kinase inhibitor therapy is a potential problem. We hypothesized that these tumors may be amenable to PD-L1 therapy given the immune nature of the tumor. PD-L1 expression in inflammatory myofibroblastic tumors has not yet been defined. The purpose of this study was to explore PD-L1 expression in inflammatory myofibroblastic tumors, as adaptive PD-L1 expression is known to enrich for response to anti-PD-1/PD-L1 therapies. Expression of PD-L1 (clone SP142) was assessed in 35 specimens from 28 patients. Positivity was defined as membranous expression in ≥5% of cells and evaluated separately in tumor and immune cells. Adaptive vs. constitutive patterns of tumor cell PD-L1 expression were assessed. PD-L1 status was correlated with clinicopathologic features. CD8+ T cell infiltrates were quantified by digital image analysis. ALK status was assessed by immunohistochemistry and/or FISH. Twenty-four (69%) tumors had PD-L1(+) tumor cells and 28 (80%) showed PD-L1(+) immune cells. Most recurrent and metastatic tumors (80%) and ALK(−) tumors (88%) were PD-L1(+). Adaptive PD-L1 expression was present in 23 (96%) of PD-L1(+) tumors, which also showed a three–four fold increase in CD8+ T cell infiltration relative to PD-L1(−) tumors. Constitutive PD-L1 expression was associated with larger tumor size (p = 0.002). Inflammatory myofibroblastic tumors show frequent constitutive and adaptive PD-L1 expression, the latter of which is thought to be predictive of response to anti-PD-1. These data support further investigation into PD-1/PD-L1 blockade in this tumor type.
- Published
- 2017
49. PD-L1 Expression in Melanoma: A Quantitative Immunohistochemical Antibody Comparison
- Author
-
Haiying Xu, Janis M. Taube, Evan J. Lipson, Sneha Berry, Genevieve J. Kaunitz, Jessica Esandrio, Karen B. Bleich, Aleksandra Ogurtsova, Joel C. Sunshine, Robert A. Anders, Tricia R. Cottrell, Toby C. Cornish, and Peter Nguyen
- Subjects
0301 basic medicine ,Cancer Research ,Sensitivity and Specificity ,Article ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,medicine ,Biomarkers, Tumor ,Humans ,Melanoma ,Tumor microenvironment ,Pathology, Clinical ,biology ,Cancer ,Antibodies, Monoclonal ,Reproducibility of Results ,medicine.disease ,Molecular biology ,Immunohistochemistry ,Staining ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Monoclonal ,biology.protein ,Antibody - Abstract
Purpose: PD-L1 expression in the pretreatment tumor microenvironment enriches for response to anti-PD-1/PD-L1 therapies. The purpose of this study was to quantitatively compare the performance of five monoclonal anti-PD-L1 antibodies used in recent landmark publications. Experimental Design: PD-L1 IHC was performed on 34 formalin-fixed paraffin-embedded archival melanoma samples using the 5H1, SP142, 28-8, 22C3, and SP263 clones. The percentage of total cells (including melanocytes and immune cells) demonstrating cell surface PD-L1 staining, as well as intensity measurements/H-scores, were assessed for each melanoma specimen using a computer-assisted platform. Staining properties were compared between antibodies. Results: Strong correlations were observed between the percentage of PD-L1(+) cells across all clones studied (R2 = 0.81–0.96). When present, discordant results were attributable to geographic heterogeneity of the melanoma tissue section rather than differences in PD-L1 antibody staining characteristics. PD-L1 intensity/H-scores strongly correlated with percentage of PD-L1(+) cells (R2 > 0.78, all clones). Conclusions: The 5H1, SP142, 28-8, 22C3, and SP263 clones all demonstrated similar performance characteristics when used in a standardized IHC assay on melanoma specimens. Reported differences in PD-L1 IHC assays using these antibodies are thus most likely due to assay characteristics beyond the antibody itself. Our findings also argue against the inclusion of an intensity/H-score in chromogenic PD-L1 IHC assays. Clin Cancer Res; 23(16); 4938–44. ©2017 AACR.
- Published
- 2017
50. Association of HIV Status With Local Immune Response to Anal Squamous Cell Carcinoma: Implications for Immunotherapy
- Author
-
Aleksandra Ogurtsova, Jessica Esandrio, Maria Libera Ascierto, Elizabeth L. Yanik, Haiying Xu, Suzanne L. Topalian, Janis M. Taube, Genevieve J. Kaunitz, Eric A. Engels, Farah Succaria, Tracee L. McMiller, Toby C. Cornish, Tricia R. Cottrell, and Evan J. Lipson
- Subjects
0301 basic medicine ,Oncology ,Adult ,Male ,Cancer Research ,medicine.medical_specialty ,Cell cycle checkpoint ,CD3 Complex ,medicine.medical_treatment ,CD8 Antigens ,T-Lymphocytes ,Antigens, Differentiation, Myelomonocytic ,HIV Infections ,B7-H1 Antigen ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Lymphocytes, Tumor-Infiltrating ,Antigens, CD ,Internal medicine ,medicine ,Tumor Microenvironment ,Humans ,business.industry ,Anal Squamous Cell Carcinoma ,Interleukin-18 ,Immunotherapy ,Middle Aged ,Anus Neoplasms ,Immunohistochemistry ,Lymphocyte Activation Gene 3 Protein ,Immune checkpoint ,Gene expression profiling ,030104 developmental biology ,030220 oncology & carcinogenesis ,Case-Control Studies ,Immunology ,CD4 Antigens ,Carcinoma, Squamous Cell ,Female ,business ,CD8 - Abstract
Importance The programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) pathway play an important immunosuppressive role in cancer and chronic viral infection, and have been effectively targeted in cancer therapy. Anal squamous cell carcinoma (SCC) is associated with both human papillomavirus and HIV infection. To date, patients with HIV have been excluded from most trials of immune checkpoint blocking agents, such as anti–PD-1 and anti–PD-L1, because it was assumed that their antitumor immunity was compromised compared with immunocompetent patients. Objective To compare the local tumor immune microenvironment (TME) in anal SCCs from HIV-positive and HIV-negative patients. Design, Setting, and Participants Anal SCC tumor specimens derived from the AIDS and Cancer Specimen Resource (National Cancer Institute) and Johns Hopkins Hospital included specimens. Tumors were subjected to immunohistochemical analysis for immune checkpoints (PD-L1, PD-1, LAG-3) and immune cell (IC) subsets (CD3, CD4, CD8, CD68). Expression profiling for immune-related genes was performed on select HIV-positive and HIV-negative cases in PD-L1 + tumor areas associated with ICs. Main Outcomes and Measures Programmed death-ligand 1 expression on tumor cells and ICs, PD-L1 patterns (adaptive vs constitutive), degree of IC infiltration, quantified densities of IC subsets, and gene expression profiles in anal SCCs from HIV-positive vs HIV-negative patients. Results Approximately half of 40 tumor specimens from 23 HIV-positive and 17 HIV-negative patients (29 men and 11 women; mean [SD] age, 51 [9.9] years) demonstrated tumor cell PD-L1 expression, regardless of HIV status. Median IC densities were not significantly decreased in HIV-associated tumors for any cellular subset studied. Both adaptive (IC-associated) and constitutive PD-L1 expression patterns were observed. Immune cell PD-L1 expression correlated with increasing intensity of IC infiltration ( r = 0.52; 95% CI, 0.26-0.78; P + T-cell density ( r = 0.35; 95% CI, 0.11-0.59; P = .03). Gene expression profiling revealed comparable levels of IFNG in the TME of both HIV-positive and HIV-negative patients. A significant increase in IL18 expression levels was observed in HIV-associated anal SCCs (fold change, 12.69; P Conclusions and Relevance HIV status does not correlate with the degree or composition of IC infiltration or PD-L1 expression in anal SCC. These findings demonstrate an immune-reactive TME in anal SCCs from HIV-positive patients and support clinical investigations of PD-1/PD-L1 checkpoint blockade in anal SCC, irrespective of patient HIV status.
- Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.