GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2988-2988
    Abstract: Background: Response to checkpoint blockade may be dependent on tumor mutational load and the presence of antigen-specific effector T cells in the tumor microenvironment; however, how blockade modulates these features during therapy is unclear. We assessed genomic changes in tumors from patients (pts) with advanced melanoma receiving nivolumab (nivo) who progressed on ipilimumab (ipi-P) or were ipi-naive (ipi-N). Methods: Tumor biopsies were collected pretreatment and 4 weeks post first nivo dose from ipi-N or ipi-P pts treated with nivo 3 mg/kg Q2W in the phase 1 open-label CA209-038 study (NCT01621490). Biopsies from 68 pts were analyzed by whole exome, transcriptome, and/or TCR sequencing (paired biopsies from 41, 42, and 34 pts, respectively). Results: Objective response rate (ORR) in the overall cohort (n=85) was 27% with similar ORR in ipi-N and ipi-P cohorts. In the genomic cohort (n=68), ORR was 23% with a similar number of complete or partial responses (CR/PR) in ipi-N and ipi-P pts (n=7 and n=8, respectively). Prior to treatment, mutational and neoantigen load were comparable, regardless of previous treatment. Following nivo treatment, both mutational and neoantigen load were reduced 5-fold in pts who responded (CR/PR; n=9) and 1.2-fold in pts with stable disease (SD, n=13) compared with a 1.1-fold increase in pts with progressive disease (PD, n=19). Intratumoral heterogeneity analysis before and after nivo demonstrated that CR/PR pts generally lost tumor mutation clones/subclones. Novel tumor mutation clones were observed in on-treatment samples from 2 CR/PR pts and all pts who progressed on nivo. Transcriptome analyses revealed significant increases in distinct tumor immune cell subsets (CD8+ T cells and NK cells) and immune checkpoint gene expression (LAG3, CTLA4, PCDC1, and CD274 [PD-L1]) following nivo, which were more pronounced in pts with CR/PR vs PD (log2 fold-changes of 1.24, 1.07, 1.71, and 0.74, respectively). Consistent with the transcriptome analyses, tumor-infiltrating lymphocytes, as assessed by immunohistochemistry, generally increased following nivo in pts who responded: 2.8 vs 1.9-fold change in CR/PR/SD vs PD in the ipi-P cohort; 4.8 vs 1.8-fold change in CR/PR/SD vs PD in the ipi-N cohort. Differences in treatment-related TCR repertoire diversity changes were apparent between pts who responded within the ipi-N and ipi-P cohorts: a decrease in the evenness of T-cell clonotype distribution was observed among pts with CR/PR/SD relative to pts with PD in the ipi-N cohort (P=0.036), but not in the ipi-P cohort. Conclusion: Nivo and ipi modulate T-cell repertoire and tumor mutational heterogeneity in pts with advanced melanoma, presenting potential mechanisms of action underlying successful nivo therapy. These data also show that prior ipi treatment may influence biological response to nivo, but further investigation is warranted. Citation Format: Timothy A. Chan, Nadeem Riaz, Jonathan J. Havel, Vladimir Makarov, Alexis Desrichard, Jennifer S. Sims, F. Stephen Hodi, Salvador Martín-Algarra, William H. Sharfman, Shailender Bhatia, Wen-Jen Hwu, Thomas F. Gajewski, Craig L. Slingluff, Sviatoslav M. Kendall, Han Chang, John-William Sidhom, Jonathan P. Schneck, Nils Weinhold, Christine E. Horak, Walter J. Urba. Immunogenomic analyses of tumor cells and microenvironment in patients with advanced melanoma before and after treatment with nivolumab [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2988. doi:10.1158/1538-7445.AM2017-2988
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cell, Elsevier BV, Vol. 171, No. 4 ( 2017-11), p. 934-949.e16
    Type of Medium: Online Resource
    ISSN: 0092-8674
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2017
    detail.hit.zdb_id: 187009-9
    detail.hit.zdb_id: 2001951-8
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Immunology Research Vol. 6, No. 2 ( 2018-02-01), p. 151-162
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 6, No. 2 ( 2018-02-01), p. 151-162
    Abstract: Despite a dramatic increase in T-cell receptor (TCR) sequencing, few approaches biologically parse the data in a fashion that both helps yield new information about immune responses and may guide immunotherapeutic interventions. To address this issue, we developed a method, ImmunoMap, that utilizes a sequence analysis approach inspired by phylogenetics to examine TCR repertoire relatedness. ImmunoMap analysis of the CD8 T-cell response to self-antigen (Kb-TRP2) or to a model foreign antigen (Kb-SIY) in naïve and tumor-bearing B6 mice showed differences in the T-cell repertoire of self- versus foreign antigen-specific responses, potentially reflecting immune pressure by the tumor, and also detected lymphoid organ–specific differences in TCR repertoires. When ImmunoMap was used to analyze clinical trial data of tumor-infiltrating lymphocytes from patients being treated with anti–PD-1, ImmunoMap, but not standard TCR sequence analyses, revealed a clinically predicative signature in pre- and posttherapy samples. Cancer Immunol Res; 6(2); 151–62. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Chemical Society (ACS) ; 2017
    In:  ACS Nano Vol. 11, No. 6 ( 2017-06-27), p. 5417-5429
    In: ACS Nano, American Chemical Society (ACS), Vol. 11, No. 6 ( 2017-06-27), p. 5417-5429
    Type of Medium: Online Resource
    ISSN: 1936-0851 , 1936-086X
    Language: English
    Publisher: American Chemical Society (ACS)
    Publication Date: 2017
    detail.hit.zdb_id: 2383064-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 976-976
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 976-976
    Abstract: Background: There has been a dramatic increase in T-cell Receptor (TCR) sequencing spurred, in part, by the clinical demand in Immuno-oncology and technological advances in TCR sequencing. However, there has been little in the way of approaches to parse the data in a biologically meaningful fashion. The ability to parse this data to understand the T-cell repertoire in a structurally relevant manner has the potential to open new discoveries about how the immune system responds to insults such as cancer and infectious diseases. Methods: Here we describe a novel method to visualize and quantify TCR repertoire sequence diversity. This method includes metrics such as visualization of repertoire with: 1) weighted phylogenetic trees that display relatedness and frequency of the sequences; 2) dominant motif analyses identifying clusters of highly homologous sequences that contribute significantly to response and; 3) TCR diversity score measuring the average relatedness (by sequence homology) of all TCR’s in a sample. To demonstrate the power of the approach, we have applied it to understanding the CD8 T Cell response to model self (TRP2) and foreign (SIY) antigens in naïve and tumor-bearing (B16 melanoma) B6 mice. Additionally, this method was applied to tumor infiltrating lymphocytes, TIL, taken pre- and on-therapy, from patients undergoing Nivolumab (α-PD1) therapy for metastatic melanoma. Results: Analysis of the naïve CD8 response demonstrated a highly conserved (measured by the TCR diversity score) and less clonal response to SIY whereas the response to TRP2 was less conserved and highly clonal. Dominant motif analysis demonstrated highly rich motifs consisting of many homologous sequences in the SIY response but few sequences per motif in the TRP2 response. This may reflect the outcome of tolerance mechanisms to self-antigens. Presence of tumor demonstrated differential immune pressure on the TRP2 vs SIY response. Tumor primed novel SIY motifs but constricted the number of dominant motifs in the TRP2 response while additionally altering the sequence of the motifs. In patients undergoing α-PD1 therapy, we identified signatures in pre- and post-therapy TCR repertoires that correlated with clinical outcome response. Prior to therapy, patients whose dominant motifs were rich with many sequences responded favorably to checkpoint inhibition over those with less rich motifs. After four week on therapy, patients whose TCR repertoires became more conserved responded more favorably to PD1 treatment while those who did not respond had no change in their TCR diversity score. Conclusions: In summary, we have developed and demonstrated a novel method to meaningfully parse and interpret TCR repertoire data and have applied it to yield a novel insight of CD8 T Cell responses to different types of antigens in model systems as well as key characteristics of TIL repertoires from patients who respond clinically to α-PD1 therapy. Citation Format: John-William Sidhom, Catherine A. Bessell, Jonathan J. Havel, Timothy A. Chan, Jonathan P. Schneck. ImmunoMap: a novel bioinformatics tool for immune cell repertoire analysis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 976. doi:10.1158/1538-7445.AM2017-976
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...