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: Infection and Immunity, American Society for Microbiology, Vol. 89, No. 12 ( 2021-11-16)
    Abstract: Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer-related death in the United States, with few effective treatments available and only 10% of those diagnosed surviving 5 years. Although immunotherapeutics is a growing field of study in cancer biology, there has been little progress in its use for the treatment of pancreatic cancer. Pancreatic cancer is considered a nonimmunogenic tumor because the tumor microenvironment does not easily allow for the immune system, even when stimulated, to attack the cancer. Infection with the protozoan parasite Toxoplasma gondii has been shown to enhance the immune response to clear cancer tumors. A subset of T. gondii proteins called soluble Toxoplasma antigen (STAg) contains an immunodominant protein called profilin. Both STAg and profilin have been shown to stimulate an immune response that reduces viral, bacterial, and parasitic burdens. Here, we use STAg and profilin to treat pancreatic cancer in a KPC mouse-derived allograft murine model. These mice exhibit pancreatic cancer with both Kras and P53 mutations as subcutaneous tumors. Pancreatic cancer tumors in C57BL/6J mice with a wild-type background showed a significant response to treatment with either profilin or STAg, exhibiting a decrease in tumor volume accompanied by an influx of CD4 + and CD8 + T cells into the tumors. Both IFN-γ −/− mice and Batf3 −/− mice, which lack conventional dendritic cells, failed to show significant decreases in tumor volumes when treated. These results indicate that gamma interferon (IFN-γ) and dendritic cells may play critical roles in the immune response necessary to treat pancreatic cancer.
    Type of Medium: Online Resource
    ISSN: 0019-9567 , 1098-5522
    RVK:
    Language: English
    Publisher: American Society for Microbiology
    Publication Date: 2021
    detail.hit.zdb_id: 1483247-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 2734-2734
    Abstract: Background: The incidence of colorectal cancer (CRC) in those of pre-screening age continues to rise. A deeper understanding of the differences in tumor biology in early age onset (EAO) cancers is needed. Differences in the tumor microenvironment have been understudied in this setting. We previously have identified versican (VCAN), a large matrix proteoglycan, as an important factor in lymphocyte exclusion in CRC. Here we explore the impact of the mutation profile and VCAN status on the infiltration of CD8+ and CD4+ lymphocytes in EAO CRC. Methods: Cancer tissues from 153 patients with CRC were stained via immunohistochemistry for CD4, CD8, and VCAN. CD4 and CD8 stains were quantified by number of positive-staining tumor-infiltrating lymphocytes per high powered field (TILs/HPF). VCAN stains were quantified on an intensity scale from 0-3+. 121 of these patients had tissue that was sequenced using the Qiagen Comprehensive Cancer targeted sequencing panel, and variations were called using Strelka. Patients were split by age into early-onset (EAO; age at diagnosis & lt;50) and later-onset (LAO; age at diagnosis 50 or later). Results: Mutations in BRAF and APC were significantly more common in LAO cancers (p & lt;0.05 for both). TP53 mutations were correlated with significantly lower CD4+ and CD8+ TILs/HPF in the LAO cohort (12.6 CD4+ TILs/HPF for TP53-wild type [WT] vs 3.5 for TP53-mutant, p & lt;0.001; 10.0 CD8+ TILs/HPF for WT vs 3.2 for mutant, p & lt;0.01), but trended opposite, though not statistically significant, in the EAO group (2.4 vs 5.9 CD4+ TILs/HPF; 3.0 vs 5.9 CD8+ TILs/HPF). PIK3CA mutations were correlated with an increase in CD8+ TILs in the LAO cohort (5.3 vs 11.9 CD8+ TILs/HPF; p & lt;0.05) but a decrease in CD4+ TILs in the EAO cohort (4.9 vs 0.8 CD4+ TILs/HPF; p & lt;0.05). TP53-WT tumors are disproportionately low in VCAN in the LAO cohort (p & lt;0.01). Differences in VCAN accumulation could account for the changes in CD8+ and CD4+ T lymphocyte infiltration across the age cohorts and TP53 mutation status. Conclusions: The impacts of TP53 and PIK3CA mutations on immune infiltration in CRC differ in EAO compared to later onset cancers, indicating that the processes controlling immune infiltration vary between age cohorts. Such differences in immune infiltration may be linked to changes in microenvironmental factors such as VCAN. These data warrant further investigation into the relationship between tumor microenvironment and mutation profiles in EAO CRC. Citation Format: Katherine A. Johnson, Philip B. Emmerich, Anna L. Lippert, Cheri A. Pasch, Linda Clipson, Wei Zhang, Kristina A. Matkowskyj, Dustin A. Deming. Impact of the mutation profile and versican status on lymphocyte infiltration in early age onset colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2734.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3533-3533
    Abstract: Background: Cancer-associated fibroblasts (CAFs) are major regulators of the immune microenvironment and therapeutic response in colorectal cancer (CRC). Neutralizing their role in modulating the immune landscape could be the key to enhancing immunotherapy success. Imatinib, dasatinib, and nilotinib are tyrosine kinase inhibitors with several kinase targets. Methods: Tissue microarrays spanning 153 patients were stained for αSMA, TAGLN, PDPN, ICAM1, and CD8. CD8 stains were quantified as number of tumor infiltrating lymphocytes per high powered field (TILs/HPF) in the epithelial compartment. All other stains were quantified by intensity on a 0-3+ scale. Scores for αSMA and TAGLN were combined into a myCAF score, and PDPN and ICAM1 into an iCAF score. myCAF gene expression signatures derived from a re-analysis of scRNA-seq data previously done by our lab were entered into the LINCS database to discover potential drugs to reverse the phenotype. Primary cancer associated fibroblasts were derived from patient tumor samples, then treated with clinically relevant concentrations of imatinib, dasatinib, or nilotinib for 96 hours. RNA was isolated and RT-qPCR was performed to quantify the myCAF genes ACTA2, COL11A1, and TAGLN, and the iCAF genes ICAM1, PDPN, IL1R1, CXCL1 and CXCL2. TGFB1 expression was also measured. Expression was normalized to untreated cells and GAPDH expression levels. Results: Cancers with high expression of myCAF markers but low expression of iCAF markers had the most CD8+ TILs (average 10.2; median 1.5; range 0-73), while cancers with low myCAF scores and high iCAF scores had the least (average 1.5; median 0; range 0-19; p & lt; 0.01). Reversing the myCAF signature relative to iCAFs in the LINCs database revealed nilotinib as a top hit. Treatment with imatinib did not significantly alter the expression of myCAF genes (control vs. max dose: p = 0.06 for ACTA2, COL11A1 p =0.2, TAGLN p = 1), while treatment with dasatinib significantly increased these genes (ACTA2 1.4x higher, p & lt; 0.001; COL11A1 2.6x higher, p & lt; 0.01; TAGLN 1.5x higher, p & lt; 0.001). Only treatment with nilotinib significantly decreased myCAF genes (ACTA2 2.2x lower, p & lt;0.001; COL11A1 1.3x lower, p =0.05; TAGLN 1.9x lower, p & lt; 0.01). All three drugs decreased iCAF gene CXCL1, and all but dasatinib decreased CXCL2. All three drugs significantly decreased TGFB1, a potential functional marker for altering myCAF phenotype (dasatinib 1.1x lower, p = 0.1; imatinib 1.6x lower, p & lt; 0.001; nilotinib 1.5x lower, p & lt; 0.05). Conclusions: myCAFs may be major actors in immune exclusion in the microenvironment, and the reversal of the myCAF phenotype may be a target for treatment with immunotherapy. Nilotinib, but not imatinib or dasatinib, is effective at decreasing expression of myCAF genes. Further research is warranted into the mechanisms of this drug on altering expression and whether these trends continue in vivo. Citation Format: Katherine Anne Johnson, Anna L. Lippert, Sean G. Kraus, Grace E. McGrath, Philip B. Emmerich, Cheri A. Pasch, Linda Clipson, Kristina A. Matkowskyj, Wei Zhang, Dustin A. Deming. Effects of tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib on cancer-associated fibroblast phenotypes in colorectal cancer [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 3533.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5102-5102
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5102-5102
    Abstract: Background: Pancreatic ductal adenocarcinoma (PDAC) recently became the third-deadliest cancer due to its resistance to effective therapies. The tumor microenvironment (TME) in PDAC is thought to contribute to this resistance, with up to 80% of the tumor bulk consisting of stroma including cancer associated fibroblasts (CAFs) and tumor-associated macrophages (TAMs). Gemcitabine (gem) and nab-paclitaxel (nab-p) is standard of care for PDAC with modest efficacy. Immunotherapies, effective in other cancers, are often combined with chemotherapy in an attempt to augment tumor response. We investigated the impact of gem and nab-p on the TME and antigen presentation of PDAC. Methods: A cancer organoid line from KPC (LSL-KrasG12D/+;LSL-Trp53R172H/+;Pdx-1-Cre) mice, was used as a murine model of PDAC. The organoids were treated in vitro with 50uM gem and 5.85uM nab-p for 12-24 hrs and subsequently tested for MHC class I expression via flow cytometry. A co-culture in Matrigel was created using KPC organoids, pro-tumor M2-polarized bone-marrow derived macrophages (BMDMs), and pancreatic stellate cells (PSCs). After 48 hrs the co-culture was treated with gem and nab-p for 24 hrs and collected to assess macrophage and fibroblast polarization markers via flow cytometry. KPC organoids were used to generate allografts in wild-type B6 mice randomized to receive 100 mg/kg gem (i.p.) and 30 mg/kg nab-p (i.v.) or control. After 96 hrs, tumors were processed for flow cytometry analyses. Results: The percentage of MHC I expressing KPC cells increased from baseline following 12 hrs of gem/nab-p treatment (32.7% vs 50.4%, p= 0.12 and declined at 24, 48 and 72 hour post-treatment time points (22.5%, p=0.3, 23.2%, p=0.31, and 13.1%, p=0.06, respectively). In the co-culture, gem and gem/nab-p significantly enhanced immunoregulatory inflammatory CAF (iCAF) subtype over control (17.5 vs 17.6 vs 10%, respectively, p & lt;0.001). A decrease in myofibroblastic CAFs (myCAFs) was also observed. M1 macrophages in the co-culture decreased significantly with gem and gem/nab-p compared to controls (6.7% vs 6.9% vs 13.8%, respectively, p=0.02/p=0.004). There was also a subsequent increase in M2 macrophages. KPC allografts demonstrated no significant change in MHC I levels following a 96-hour gem/nab-p treatment compared to vehicle. The CAF subtypes showed trends similar to the in vitro co-culture, with more iCAFs (14.9% vs 26.3%, p=0.32) and less myCAFs (83.5% vs 71.3%, p=0.31). We were also able to identify an MHC II class of antigen-presenting CAFs (apCAFs), within both our in vitro and in vivo studies at levels less than 1% of all CAFs. However, no change was observed with treatment. Conclusion: Here we demonstrate reduced MHC class I expression, increased M2 TAM polarization, and increased iCAFs, all associated with an immunoregulatory phenotype. These finding suggest that further studies on alternative combinations of immunotherapies are warranted. Citation Format: Hanna R. Rainiero, Philip B. Emmerich, Nathaniel Verhagen, Rebecca Destefanis, Cheri A. Pasch, Linda Clipson, Kristina A. Matkowskyj, Dustin A. Deming. Gemcitabine and nab-paclitaxel increase immunosuppressive environment in PDAC [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 5102.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    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 ...
  • 5
    In: The Journal of Immunology, The American Association of Immunologists, Vol. 199, No. 5 ( 2017-09-01), p. 1933-1941
    Abstract: Colorectal cancer originates within immunologically complex microenvironments. To date, the benefits of immunotherapy have been modest, except in neoantigen-laden mismatch repair–deficient tumors. Approaches to enhance tumor-infiltrating lymphocytes in the tumor bed may substantially augment clinical immunotherapy responses. In this article, we report that proteolysis of the tolerogenic matrix proteoglycan versican (VCAN) strongly correlated with CD8+ T cell infiltration in colorectal cancer, regardless of mismatch repair status. Tumors displaying active VCAN proteolysis and low total VCAN were associated with robust (10-fold) CD8+ T cell infiltration. Tumor-intrinsic WNT pathway activation was associated with CD8+ T cell exclusion and VCAN accumulation. In addition to regulating VCAN levels at the tumor site, VCAN proteolysis results in the generation of bioactive fragments with novel functions (VCAN-derived matrikines). Versikine, a VCAN-derived matrikine, enhanced the generation of CD103+CD11chiMHCIIhi conventional dendritic cells (cDCs) from Flt3L-mobilized primary bone marrow–derived progenitors, suggesting that VCAN proteolysis may promote differentiation of tumor-seeding DC precursors toward IRF8- and BATF3-expressing cDCs. Intratumoral BATF3-dependent DCs are critical determinants for T cell antitumor immunity, effector T cell trafficking to the tumor site, and response to immunotherapies. Our findings provide a rationale for testing VCAN proteolysis as a predictive and/or prognostic immune biomarker and VCAN-derived matrikines as novel immunotherapy agents.
    Type of Medium: Online Resource
    ISSN: 0022-1767 , 1550-6606
    RVK:
    RVK:
    Language: English
    Publisher: The American Association of Immunologists
    Publication Date: 2017
    detail.hit.zdb_id: 1475085-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2019
    In:  Current Colorectal Cancer Reports Vol. 15, No. 2 ( 2019-4), p. 61-69
    In: Current Colorectal Cancer Reports, Springer Science and Business Media LLC, Vol. 15, No. 2 ( 2019-4), p. 61-69
    Type of Medium: Online Resource
    ISSN: 1556-3790 , 1556-3804
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2233562-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4861-4861
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4861-4861
    Abstract: Introduction: Immunotherapies are increasingly being used for patients diagnosed with cancer, however, for metastatic colorectal cancer (CRC), more than 95% of patients have shown little to no clinical benefit to immunotherapy. Previous work from our lab has shown that copanlisib, a PI3K inhibitor, was found to enhance MHC class I expression in Kras mutant CT26 murine colon cancer cells. Here we examine the potential for anti-cancer activity with the combination of copanlisib and anti-PD1 treatments using this model. Methods: The impact of copanlisib on CT26 cells was performed using the WST assay and the expression of MHC class I was assessed using flow cytometry. CT26 flank allografts were generated in Balb/C mice and subsequently treated for 15 days with copanlisib (10 mg/kg), anti-PD1 (0.2 mg, BioXCell) or the combination. An IgG2a antibody (BioXCell) was used as a control. Tumors were measured twice a week using a caliper. Tumors were excised, and prepared for immunohistochemistry for CD8, CD4, perforin, granzyme B, and F480 was performed. Staining was quantified as the number of positive staining cells per 20X field of view (FOV). Results: CT26 viability in response to 200 nM copanlisib was relatively unchanged as compared with untreated controls. Via flow cytometry, a 75% increase in mean fluorescent intensity (MFI) for MHC class I was observed comparing control to copanlisib (p=0.003). In vivo, after 15 days there was no difference in the growth rate of those cancers treated with control versus copanlisib or anti-PD1. A significant reduction in growth rate was observed with the combination of anti-PD1 with copanlisib compared to the other arms (median fold change=3.62; control: 7.49, p=0.002, copanlisib: 8.88, p=0.003; anti-PD1: 8.93, p=0.002). There were no differences in CD8 T cell and perforin expression between the treatment groups. Granzyme B expression was higher in the combination compared (median/FOV= 44) to the control (27, p= 0.01). Additionally, a significant reduction in F480 expression was seen in the combination compared to the other treatment groups (median/FOV=43; control: 139, p & lt;0.001, copanlisib: 110, p & lt;0.001; anti-PD1:149, p & lt;0.001). Conclusions: In conclusion, copanlisib in combination with anti-PD1 demonstrated enhanced anti-tumor activity in Balb/c mice that were injected with CT26 CRC cells. This response was correlated with increased granzyme B expression and a reduction in macrophages with the combination treatment. Further studies will expand on the mechanism of this combination therapy. Citation Format: Alexa E. Schmitz, Kennedy J. Maduscha, Sarbjeet Makkar, Cheri A. Pasch, Rebecca A. DeStefanis, Philip B. Emmerich, Dustin A. Deming. Copanlisib enhances the effectiveness of anti-PD1 therapies for colorectal cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4861.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 3166-3166
    Abstract: Background: Incidences of early age onset colorectal cancer (CRC) (those diagnosed before age 50) have been increasing by 2% every year since the 1990s, with the rate expecting to double by 2030. Although there has been some research done on differences in mutational profiles between patients diagnosed before and after age 50, little research has been done to understand the immune and stromal environments of early age onset (EAO) CRC. As identified by previous literature, there are two distinct phenotypes of cancer associated fibroblasts (CAFs): myofibroblastic (myCAFs) and non-myofibroblastic (non-myCAFs). Here, we evaluate both CAF environments as well as immune infiltrating cells in context of EAO CRC. Methods: A total of 153 CRC patient samples were obtained with matching adjacent normal tissue. Of these, 60 patients had EAO CRC. Tissue slides were stained via immunohistochemistry (IHC) for the CAF subtype markers αSMA, FAP, PDPN, and MMP2, by Masson's Trichrome for collagen, and then quantified on an intensity scale from 0-3. MyCAF and non-myCAF scores were calculated by averaging the scores of αSMA and collagen, or FAP, PDPN, and MMP2, respectively. Once these scores were determined, they were split into low (average score & lt;2) and high (average score ≥2) groups. CD4 and CD8 IHC stains were quantified as the number of tumor infiltrating lymphocytes (TILs) per high power field (HPF) in the epithelial compartment. Results: Cancers with a low myCAF and non-myCAF score display the highest average number of CD4+ (10.6) and CD8+ (10.3) TILs across both age groups. Furthermore, cancers with both myCAF and non-myCAF high scores had reduced average CD4+ and CD8+ TILs when compared to both CAF scores being low (p value: 0.018 for CD8+ TILs, & lt;0.001 for CD4+ TILs). Also, cancers that show myCAF high and non-myCAF low scores show the overall lowest average CD8+ TILs. Comparing the age groups directly, there are significantly higher CD4+ TILs in the 50+ age group in all CAF phenotypes except non-myCAF high (p-values 0.01 myCAF low, 0.05 myCAF high, 0.007 non-myCAF low, 0.36 non-myCAF high). However, for CD8+ TILs, the EAO CRC group trends towards higher CD8+ TILs in the myCAF high (3.7 vs 2.1) and non-myCAF high cancers (5.7 vs 3.3), but lower CD8+ TILs in myCAF low (6.8 vs 10.5) and non-myCAF low cancers (4.0 vs 8.4) when compared to the 50+ age group. Conclusions: Here we demonstrate that there are differences in the stromal and immune microenvironments between both age groups of CRC. We indicate that increased myCAF and non-myCAF scores are associated with T cell exclusion. However, the extent of T cell exclusion is different in EAO CRC compared to patients diagnosed after age 50. The reasoning for the difference in T-cell exclusion remains unknown and this implicates the importance for further research into the stromal and immune microenvironments of EAO CRC. Citation Format: Anna Lippert, Katherine A. Johnson, Philip B. Emmerich, Cheri A. Pasch, Linda Clipson, Kristina A. Matkowskyi, Wei Zhang, Dustin A. Deming. Impact of cancer associated fibroblast phenotypes on the infiltration of t-lymphocytes in early age onset colorectal cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 3166.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 3198-3198
    Abstract: Background: Cancer Associated Fibroblasts (CAFs) are a significant component of tumor stroma, and have an important impact on immune infiltration in the tumor microenvironment (TME). Two major subtypes of CAFs have been previously identified by literature: myofibroblastic (myCAF) and inflammatory (iCAF). Our lab has identified subtype markers for each CAF phenotype and previously analyzed a sampling of 153 colorectal cancer (CRC) patients. Here, we validate these subtype markers and investigate CAF phenotypes in metastatic colorectal cancer patients. Methods: Dual immunofluorescence on formalin fixed paraffin embedded tissue sections was performed to analyze co-staining between combinations of myCAF markers, αSMA and TAGLN, and iCAF markers, PDPN and ICAM1. Slides were imaged using a fluorescent microscope. Also, tissue microarrays sampling 212 CRC patients spanning all stages of disease, 90 with matched metastatic cores, were stained via immunohistochemistry (IHC) for the CAF subtype markers then quantified on an intensity scale from 0-3+. iCAF and myCAF marker scores were averaged to get a composite score for each, then split into low (average score & lt;2) and high (average score ≥2) groups. CD8 IHC stains were quantified as the number of tumor infiltrating lymphocytes (TILs) per high power field (HPF) in the epithelial compartment. Results: Significant co-staining was observed between iCAF markers PDPN and ICAM1, as well as myCAF markers αSMA and TAGLN. Co-staining did not occur, or was minimal, between combinations of myCAF and iCAF markers. There is not significant different in abundance of iCAFs or myCAFs in primary site cores of patients with metastatic versus non-metastatic disease (p = 0.67 for iCAF, p = 0.57 for myCAF). Of matched primary and metastatic samples able to be scored, 43.3% of samples had a decrease in iCAF score from primary to metastatic site while only 18.8% increased. Overall, 34.4% of samples had a decrease in score of more than 1 and only 2.2% of samples had an increase of more than 1. However, the percentage of samples that had a decrease in myCAF score was 32.2% while 22.2% increased. In all primary cores of patients with metastatic disease, there was higher average CD8+ TILs in those with high iCAF scores compared to those with low iCAF scores (12.0 vs 5.5, p=0.03). There was not a significant difference in average CD8+ TILs in those with high myCAF scores compared to those with low myCAF scores (9.3 vs 7.1, p=0.7). Conclusions: Here, we validate the myCAF markers TAGLN and αSMA, as well as, iCAF markers ICAM1 and PDPN by demonstrating co-staining between CAFs of the same subtype and exclusion between different subtypes. These data indicate that that CAF phenotype correlates with CD8 T cell infiltration into the TME. iCAFs correlate with immune infiltration and myCAFs with immune exclusion. Citation Format: Anna L. Lippert, Katherine A. Johnson, Cheri A. Pasch, Sean G. Kraus, Philip B. Emmerich, Linda Clipson, Kristina A. Matkowskyj, Wei Zhang, Dustin A. Deming. Validation and analysis of cancer associated fibroblast subtype markers in metastatic colorectal cancer [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 3198.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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 ...
  • 10
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4583-4583
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4583-4583
    Abstract: Introduction: Neuroendocrine tumors (NETs) are a rare, understudied form of human cancer lacking robust preclinical models. Our laboratory has recently demonstrated the importance of the tumor microenvironment (TME) in T-cell exclusion from tumors. Here we examine the accumulation of the immunoregulatory proteoglycan, versican (VCAN), and its immunostimulatory proteolysis product, versikine (Vkine), produced by VCAN cleavage via ADAMTS proteins. Immunotherapeutics are increasingly being studied in NETs and investigation of the immune permissive nature of their TME is warranted. Methods: Human NET tissue microarrays (TMAs) were obtained from the University of Wisconsin Carbone Cancer Center TSB Biobank, containing human NET tissues across a broad spectrum of disease sites including the lung and gastrointestinal (GI) tract among others. Immunohistochemistry was performed using antibodies to VCAN, Vkine, and CD8+ T-cells. VCAN and Vkine levels were measured using a four-tiered relative intensity system (0/1/2/3), while CD8+ T-cell infiltration was measured by counting the CD8+ T-cells per high power field (hpf). Experimental groups were binned as low if VCAN or Vkine scored 0 or 1 and high if VCAN or Vkine scored 2 or 3. Results: Across all samples, high VCAN levels were observed in 20% of samples while high Vkine levels were observed in 55% of samples. Within the lung NET subset, which includes small cell lung cancer, 15% of samples were designated VCAN high and 65% were Vkine high. For the GI NET subset, 34% of samples scored as VCAN high while 50% scored as Vkine high. This study identifies a greater amount of Vkine high tumors present in NETs than we previously observed for colorectal, anal, pancreatic, and breast cancers. The proteolytic predominant phenotype (VCAN low and Vkine high) was present in 55% of lung NETs and 31% of GI NETs. CD8+ T cell infiltration correlated with the VCAN proteolysis predominant phenotype (VCAN low/Vkine High - CD8+ T-cell 16/hpf vs. all other VCAN/Vkine phenotypes 6.5/hpf, p & lt;0.001). This trend persisted across all NET specimen types, including lung and GI primary NETs. Discussion: VCAN accumulation occurs in a subset of NETs, likely leading to immune exclusion and potential resistance to immunotherapies. Excitingly, we demonstrate that a large subset of these cancers has active proteolysis of VCAN in the TME leading to release of the immunostimulatory fragment Vkine. Those NETs with low levels of VCAN and high levels of Vkine are predicted to have a permissive TME for immunotherapies. Further studies are needed to discern the underlying mechanisms resulting in this proteolysis and the signaling consequences with Vkine production. Additionally, this study indicates that VCAN proteolysis should be investigated as a novel biomarker for immunotherapy response for NETs. Citation Format: Christopher P. Babiarz, Philip B. Emmerich, Carley M. Sprackling, Cheri A. Pasch, Linda Clipson, Kristina A. Matkowskyj, Fotis Asimakopoulos, Dustin A. Deming. Versican accumulation and proteolysis in neuroendocrine tumors [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4583.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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...