Virtual Library

Start Your Search

J. Botling



Author of

  • +

    MA 05 - Immuno-Oncology: Novel Biomarker Candidates (ID 658)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Immunology and Immunotherapy
    • Presentations: 1
    • +

      MA 05.13 - Scavenger Receptor MARCO Defines a Targetable Tumor-Associated Macrophage Subset in Lung Cancer (ID 8641)

      15:45 - 17:30  |  Author(s): J. Botling

      • Abstract
      • Presentation
      • Slides

      Background:
      Tumor-associated macrophages (TAMs) with immunosuppressive and tumor promoting features are attractive targets for immunotherapy. MARCO is a scavenger receptor expressed on a subpopulation of macrophages in secondary lymphoid organs. A recent study performed in animal models concluded that treatment with an anti-MARCO antibody results in reprogramming of the TAMs and inhibition of tumor growth and metastatic spread. The expression and function of MARCO in lung cancer TAMs is not known.

      Method:
      The infiltration of TAMs expressing MARCO, CD68, CD163 and MSR1, in the tumor and stromal compartments, was analyzed by immunohistochemistry in a non-small cell lung cancer (NSCLC) cohort (n=352). In addition, PD-L1 expression was assessed on tumor cells. Immunofluorescence was performed on selected cases to evaluate marker co-expression. Associations to immune cells and regulatory inflammatory pathways were studied in a subset of cases (n=174) with available RNA-seq data.

      Result:
      A large variance in TAM density could be observed between cases as well as a strong correlation between CD68 and CD163, indicating a pro-tumor phenotype of infiltrating macrophages. Correlation to clinical data showed a trend towards worse survival for patients with high macrophage infiltration. TAM expression of MARCO was seen on a subpopulation of pro-tumor macrophages. The majority of MARCO expressing TAMs were found to be located within tumor cell nests. Interestingly, stromal macrophages expressing MARCO tended to aggregate in close proximity to the tumor nests. On the transcriptomic level, increased MARCO gene expression correlated to genes linked to immunosuppressive TAMs, T-cell infiltration and immune checkpoint molecules like PD-L1 and CTLA-4. The association between macrophage infiltration and tumor cell PD-L1 expression was confirmed by immunohistochemistry. Also, co-expression of PD-L1 and MARCO could be detected on certain macrophages within the tumor cell nests.

      Conclusion:
      MARCO expression characterizes a specific subpopulation of pro-tumor macrophages that are enriched in PD-L1 positive NSCLC cases. Patients with significant infiltration of MARCO positive TAMs could benefit from treatment with anti-MARCO antibodies, possibly in combination with available immune checkpoint inhibitors.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    MA 06 - Lung Cancer Biology I (ID 660)

    • Event: WCLC 2017
    • Type: Mini Oral
    • Track: Biology/Pathology
    • Presentations: 1
    • +

      MA 06.01 - Cancer Testis Antigens and Mutational Load in Relation to the Immune Landscape of Non-Small Cell Lung Cancer (ID 9369)

      15:45 - 17:30  |  Author(s): J. Botling

      • Abstract
      • Presentation
      • Slides

      Background:
      The avoidance of immune surveillance by tumor cells is an accepted hallmark of cancer. The aim of this study was to describe the natural immune landscape of NSCLC tissue, to identify important regulatory associations and potential targets of immune response. This includes mutational load and cancer testis antigen (CTA) expression, and the comprehensive analysis of tumor infiltrating immune cells in connection with immune signaling and clinical information.

      Method:
      Tissue microarrays including duplicate cancer samples of 357 NSCLC patients were stained with antibodies against CD3, CD4, CD8, CD45RO, FoxP3, CD20, CD138, and CD44 to analyze the protein expression in the stroma and tumor compartment. For 197 of these cases, corresponding RNA-seq data were available. The immunological data were correlated to the transcriptomic data and to patients’ clinical outcome. The mutation status and the mutational load was based on a targeted next-generation sequencing panel of 82 genes (HaloPlex).

      Result:
      The immune cell infiltration was predominantly in the stroma, although CD8 and FoxP3 cells also showed relevant infiltration of the tumor cell compartment. The amount of T-cells of different subsets and CD20-positive B-cells correlated positively to each other. A higher mutational load was associated with higher CD8 T-cell infiltrates, CD45RO cells, FoxP3 regulatory cells as well as CD20-positive B-cells in the tumor compartment. In contrast, the number of expressed CTAs were associated with an abundance of CD45RO-positive cells in the stromal compartment. Only CD44-positivity (HR = 0.61, p< 0.01) as well as high CD20 positive B-cells (HR = 0.34, p< 0.01) and plasma cell (CD138, HR = 0.71, p< 0.05) counts in the tumor, and for plasma cells also the stromal (HR = 0.61, p< 0.01), compartment were associated with longer overall survival.

      Conclusion:
      Here we describe natural immune profiles in a large clinical NSCLC patient cohort. Interestingly both mutational load and CTA expression is associated with the abundance of distinct immune cell infiltrates. We could not confirm the impact of tumor infiltrating T-cells on survival. However, the consistent prognostic impact of both B-cell markers indicates a major role of the humoral immune response in lung cancer.

      Only Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login, select "Add to Cart" and proceed to checkout. If you would like to become a member of IASLC, please click here.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    P2.02 - Biology/Pathology (ID 616)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Biology/Pathology
    • Presentations: 1
    • +

      P2.02-015 - Mutation Patterns in a Swedish Non-Small Cell Lung Cancer Cohort (ID 10048)

      09:30 - 16:00  |  Author(s): J. Botling

      • Abstract
      • Slides

      Background:
      Non-small cell lung cancer (NSCLC) is a heterogeneous disease with unique combinations of somatic molecular alterations in individual patients, as well as significant differences in populations across the world with regard to mutation spectra and mutation frequencies. Here we describe the mutational pattern and linked clinical parameters in a population-based Swedish NSCLC cohort.

      Method:
      The cohort consists of 354 patients treated surgically at the University Hospital in Uppsala between 2006 to 2010. DNA was extracted from either fresh frozen (n=200) or formalin fixed paraffin embedded (FFPE; n=154) tissues prior to library preparation with Haloplex capture probes and Illumina Hiseq sequencing. The gene panel covers all exons of 82 genes, that have been shown to harbor mutations relevant for NSCLC development, and utilizes a reduced target fragment length and two strand capture compatible with degraded FFPE samples.

      Result:
      In order to avoid a systematic technical bias between FFPE and fresh-frozen samples, we adapted the sequencing depth and the bioinformatic pipeline for variant calling to obtain uniform sequence coverage and mutational load across the two sample types. TP53 was the most frequently mutated gene in both adenocarcinoma (AdC; 47%) and squamous cell carcinoma (SqC; 85%). KEAP1 or NFE2L2 was mutated in 19% of AdC and 23% of SqC in a mutually exclusive fashion. In AdC, hotspot alterations in driver genes could be seen in KRAS (43%), EGFR (13%), ERBB2 (3%, exon 20 insertions), BRAF (2%) and MET (1%, exon 14 skipping). Mutations in STK11 were observed in 21% of AdC cases. In SqC, frequently mutated genes were MLL2 (26%), PIK3CA (20%), CDKN2A (15%) and DDR2 (4%). Survival analysis revealed a worse overall survival for AdC patients with a mutation in either TP53, STK11 or SMARCA4. In the KRAS-mutated group poor survival appeared to be linked to concomitant TP53 or STK11 mutations, and not to KRAS mutation as a single aberration. In SqC a worse overall survival could be observed for patients with MLL2 mutations. SqC patients with mutations in CSMD3 had trend for a better prognosis.

      Conclusion:
      Here we have evaluated the mutational status of a Swedish NSCLC cohort. Technical adaption allowed analysis across both FFPE and fresh-frozen samples. Overall, the high frequency of TP53 and KRAS mutations might be related to the large fraction of smokers. Poor prognosis was linked to mutations in TP53, STK11 or SMARCA4 in AdC and MLL2 mutations in SqC.

      Only Active Members that have purchased this event or have registered via an access code will be able to view this content. To view this presentation, please login or select "Add to Cart" and proceed to checkout.

  • +

    P3.02 - Biology/Pathology (ID 620)

    • Event: WCLC 2017
    • Type: Poster Session with Presenters Present
    • Track: Biology/Pathology
    • Presentations: 1
    • +

      P3.02-097g - LRIG1 and LMO7 Are Interacting Proteins with Clinical Significance in NSCLC (ID 9406)

      09:30 - 16:00  |  Author(s): J. Botling

      • Abstract

      Background:
      The LRIG family of proteins consists of the paralogs LRIG1, LRIG2 and LRIG3. LRIG1 is the most studied and interacts with several receptor tyrosine kinases (RTKs), including EGFR and MET, in an inhibitory fashion. High levels of LRIG1 in tumor tissue has been associated with better clinical outcome in several solid cancers. In a previous study concerning 360 surgically treated NSCLC we showed that high LRIG1 immunoreactivity was an independent positive prognostic factor for survival. Regarding LRIG2 and LRIG3, some data indicate that they may oppose the function of LRIG1. The molecular mechanisms behind the various cellular and clinical functions of the LRIG proteins remain incompletely understood.

      Method:
      To gain further insight into the molecular functions of the LRIG proteins, we performed a yeast two-hybrid screen using a cytosolic region that is conserved in all three LRIG paralogs as the bait. Transfected cells were either lysed for immunoprecipitation, or fixated and stained for confocal microscopy using fluorescently labeled FLAG and Myc antibodies. A TMA was stained using polyclonal LRIG1 and LMO7 antibodies.

      Result:
      Preys representing LMO7 and LIMCH1 reproducibly produced viable yeast colonies, suggesting a direct interaction between LRIG1 and the two proteins. In mammalian cells, over-expressed LIMCH1 co-precipitated with both LRIG1 and LRIG3, whereas LMO7 co-precipitated with LRIG3. Confocal microscopy showed co-localization between LMO7, LIMCH1 and all three LRIG proteins. To assess the clinical significance of LMO7, the same TMA as was previously used to study the clinical significance of LRIG1 was immunostained. Positive LMO7 immunostaining did not correlate with survival in itself, even though a trend towards worse survival was seen for the positive cases. However, the previously shown survival benefit for LRIG1 high-expressing cases was limited to the LMO7 negative subgroup. This was further reinforced by a multivariable analysis, suggesting that LMO7 interacts with LRIG1 in a way that inhibits its tumor suppressive function.

      Conclusion:
      Taken together, our findings indicate a novel interaction between LRIG1 and LMO7 with clinical significance in NSCLC.