Browsing Over 2184 Presentations

Sort By Presentation Number
Basic science

4P - Spatially resolved transcriptome elucidates bidirectional tertiary lymphoid structure interacts with tumor microenvironment of non-small cell lung cancer

Presentation Number
4P
Speakers
  • Xin Zhao (Shenzhen, China)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

The presence of tertiary lymphoid structure has a significant correlation with the lung cancer patient's response to immune checkpoint inhibitors. Nonetheless, the developmental characteristics of TLS and their surrounding tumor microenvironmental features in the primary tumor have not been elucidated comprehensively.

Methods

We utilized spatially resolved transcriptome and single-cell RNA sequencing to examine the molecular signatures and the essential cell types of TLS during its maturation process in the tumor microenvironment. Additionally, multiplex immunofluorescence staining and whole transcriptome sequencing were performed for validation.

Results

We categorized 406 potential TLSs into four states: pre-TLS, primary TLS, mature TLS, and aging TLS - based on their spatial molecular characteristics. The high expression of FDCSP in follicular DCs is a novel marker to promote TLS maturation by specifically binding to activated B cells. Three types of receptor-ligand interactions in mature and aging TLS could predict the prognosis in early-stage LUAD. Interestingly, aging TLS with significantly upregulated FOXP3 and TOX2 could limit anti-tumor immunity. Additionally, the molecular characteristics of aging TLS showed a lower expression of a group of CC chemokine ligands compared to mature TLS, which could potentially lead to a weaker anti-tumor response. We also observed an increase in tumor proliferation activity in the periphery of the aging TLS region. One approach to reverse the anti-tumor activity of aging TLS to that of mature TLS could be to increase the concentration of environmental chemokines while simultaneously blocking the upregulation of TIGIT in aging TLS.

Conclusions

Those results demonstrated that the condition of TLS was influenced by the neighboring tumor microenvironment in the early-stage LUAD. And the conventional mature TLS would eventually reach a biologically aged state in the same tumor microenvironment. By regulating chemokine signaling in the aging TLS's surrounding environment, it could potentially serve as an adjuvant therapy to enhance the antitumor activity of immune elements in resectable LUAD patients.

Legal entity responsible for the study

TJMUCH and BGI.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

5P - Tertiary lymphoid structures (TLS) presence and stromal blood vessels heterogeneity differentially influence recurrence, lymphovascular, and perineural invasion in breast cancer molecular subtypes

Presentation Number
5P
Speakers
  • Andrei A. Cosma (Timisoara, Romania)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Tertiary Lymphoid Structures (TLS) mediate local antitumor immunity. Since cancer immunotherapy, TLS interests grown considerably. We examined TLS and tumor stromal blood vessels for BC molecular subtypes associated with recurrence, lymphovascu-lar (LIV), and perineural invasion (PnI).

Methods

CD34/SMA double immunostaining revealed TLS high endothelial venules and BC tumor stromal immature and mature blood vessels. Statistical study linked microscopic data to recurrence, LIV, and PnI.

Results

TLS- subgroups have higher LIV, PnI, and recurrence (p<0.001), except for Luminal A type. LIV and PnI rise in HER2+/TLS- subgroups (p<0.001). TNBC/TLS- had the highest recurrence and invasion risks. TNBC/TLS- subgroup recurrence is related to tumor grade and LIV/PnI (p<0.001), unlike other TLS- subgroups. PnI but not LVI are related with TNBC/TLS+ subgroup recurrence (p<0.001).

Conclusions

TLS affect BC molecular subtype recurrence, LVI, and PnI differently. Its antitumor immunity mediator seems closely associated to tumor aggressiveness and stromal blood vessels.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

6P - Combined single-cell and spatially resolved mapping of the human lymph node ecosystem reveals fundamental principles of lymphoma tissue organization

Presentation Number
6P
Speakers
  • Daniel Hübschmann (Heidelberg, Germany)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Lymph nodes (LN) act as central hubs for the orchestration of adaptive immune responses. A network of lymph node stromal cells (LNSC), including mesenchymal and endothelial cells, build a 3D network for immune cells to migrate and home to distinct niches in the LN. Within B cell non-Hodgkin lymphomas, indolent follicular lymphomas (FL) are characterized by subtle structural changes while the compartmentalization of the tissue remains present. In contrast, aggressive diffuse large B cell lymphomas (DLBCL) are defined by a complete loss of tissue organization indicating a potential role of LNSCs in tumorigenesis. While the morphological differences are well-known, the underlying molecular and cellular mechanisms remain poorly understood.

Methods

Here, we dissect the mechanisms underlying loss of structure in lymphomagenesis using combined single-cell transcriptome and spatially-resolved mapping approaches of LNSCs and immune cells.

Results

Using ultra-high plex immunofluorescence imaging, we characterized how lymph node cells organize into spatially distinct cellular neighborhoods, the disruption of which was congruent with lymphoma-induced remodeling. Using single-cell transcriptome data, we investigated the molecular programs driving this loss of organization. In DLBCL, chemokines relevant for immune cell organization were downregulated, while chemokines contributing to inflammation were upregulated, suggesting a phenotypic switch from a structural organized to an inflammatory and fibrotic state. In silico cell-cell interaction analysis indicated that besides the loss of mesenchyme-derived chemokine gradients, inflammatory immune cells outside of follicles turn into ectopic sources of these chemokines, an effect which likely contributes to the diffuse growth pattern of aggressive lymphomas. High expression of organizing chemokines was associated with better overall survival, this association being significant in FL and a trend in DLBCL.

Conclusions

Collectively, these data suggest that a reprogramming of the LN microenvironment triggers an imbalance in chemokine gradients, which underlies loss of tissue organization in aggressive lymphomas.

Legal entity responsible for the study

The authors.

Funding

Has not received any funding.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

7P - Engineered salmonella blocks cancer metastasis by activating NK cells in an IFN-γ-dependent manner

Presentation Number
7P
Speakers
  • JIANDONG HUANG (Hong Kong, Hong Kong PRC)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Metastasis accounts for 90% of cancer related deaths and blocking of metastatic cascade has critical clinical impact. However, the clinical drug development for cancer treatment, including cancer immunotherapies, is evaluated largely depending on their ability to cause tumour shrinkage and ignores the effect on metastasis as it has proven challenging to target. Therefore, there is an urgent need for novel therapeutic strategies and agents targeting metastasis. Using an attenuated Salmonella typhimurium strain YB1 engineered by our lab, we have found a potent suppressive effect of attenuated Salmonella on cancer metastasis, regardless of cancer types and genetic background, by evoking strong anti-metastatic immune response.

Methods

Mutant mice and antibody-mediated cell depletion were used to identify the host genetic and cellular requirement for the bacterial supression of cancer cell metastasis. CyTOF (mass cytometry or cytometry by time of flight) was used to investigate the the innate immune responses after Salmonella treatment.

Results

Our studies showed that suppression of cancer metastasis by attenuated Salmonella only requires the innate immune response. Among the many induced cytokines, IFN-γ was identified as an indispensable factor for inhibiting cancer metastasis. CyTOF and antibody-mediated cell depletion analysis of the innate immune responses after Salmonella treatment, revealed that NK cells are the major factor involved in Salmonella-provoked metastasis suppression.

Conclusions

We found that IFN-γ was mainly produced by NK cells during early Salmonella infection, and in turn, IFN-γ promoted the accumulation, activation and cytotoxicity of NK cells. The IFN-γ-dependent NK cells directly eliminated newly accumulated cancer cells in the lung to block the cancer metastasis cascade in response to the Salmonella treatment.

Legal entity responsible for the study

The author.

Funding

This work was supported by grants from the Shenzhen Peacock Team Project (KQTD2015033117210153) and Shenzhen Science and Technology Innovation Committee Basic Science Research Grant (JCYJ20170413154523577).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

8P - Modulating tumor microenvironment using a VEGF active immunotherapeutic approach in gastrointestinal tumors: Beyond angiogenesis modulation

Presentation Number
8P
Speakers
  • Mónica Bequet-Romero (Havana, Cuba)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Targeting of the vascular endothelial growth factor (VEGF) is known for going beyond mere modification of angiogenesis, and extends to the systemic and intratumoral regulation of immune cells activation state. In this study, we aim to explore the effect of a VEGF active immunotherapy based on a recombinant antigen named CIGB-247, in the systemic and tumor compartments in preclinical settings for mice, and in a subset of 22 patients with advanced gastrointestinal cancer (GIC) from phase I clinical trials (RPCEC00000102 and RPCEC00000155).

Methods

BALB/c mice were immunized with CIGB-247, and challenged with CT26 colon carcinoma cells subcutaneously. A week after the last immunization mice were euthanized and the tumor and the systemic compartment, were analyzed by ELISA, flow cytometry, and RT-QPCR assays. GIC patients’ serum and leukocyte samples were evaluated for cytokines, specific VEGF antibody titers, and VEGFR2 neutralization using ELISA, while the cellular response was assessed by ELISPOT.

Results

The vaccine significantly boosted the infiltration of activated CD3+ cells and caused a decrease in the number of MDSCs and Tregs in analyzed compartments. The levels of VEGF, PLGF, and CXCL1 were significantly reduced in the tumor lysates, while the concentration of CD8 activation-related molecules like IFN-γ, and GRZ-B were induced. Finally, we analyzed GIC cases within the two phase I clinical trials after stratification between responders and non-responders. The evaluations indicate a significant increase in the VEGF-specific humoral and cellular response parallel to a reduction in VEGF that relates significantly to increased survival. Within this small cohort, reduction or stabilization of other proangiogenic factors was also detected.

Conclusions

CIGB-247 antigen-based vaccine increased tumor-infiltrating lymphocytes and reversed the immunosuppressive effect of the tumor microenvironment in the CT26 mouse model. Such results were corroborated in a GIC patient cohort indicating that the strategy might be a successful treatment option for such patients by modifying systemic and tumor microenvironment beyond angiogenesis modulation.

Clinical trial identification

RPCEC00000102, RPCEC00000155.

Legal entity responsible for the study

Center for Genetic Engineering and Biotechnology.

Funding

HEBERBiotec.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

9P - Identification of a μCT-based radiomic signature of CD8+ tumour infiltrating lymphocytes in an orthotopic murine model

Presentation Number
9P
Speakers
  • Giulia Mazzaschi (Parma, Italy)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

In the era of immunotherapy (IT), radiomics emerged as a non-invasive tool to decode tumor immune microenvironment (TIME) and predict IT response. Intrinsic patient and tumor variability challenging the explainability of radiomic readouts might be overcome in preclinical settings. Thus, we aimed to develop a μCT radiomic platform in a CD8 T cell functionally manipulated orthotopic murine model, closely mimicking human head and neck cancer, to validate the biological reality of radiomics in assessing tumor infiltrating lymphocytes (TILs).

Methods

Set-up, training and validation experiments (N Tot = 50 C57BL/6 mice) were conducted as follows: submucosal injection of 0.5 x 106 TC1-luc cells in the right inner lip and generation of an orthotopic model of head and neck cancer (day [D] -7); tailored treatment consisting of irradiation [D0] ± anti-CD8 antibody [D3] to selectively enrich or deplete CD8+ TILs; in vivo preclinical imaging through Quantum FX μCT technology [D3, D4]; mice euthanasia and tumor sampling [D4], followed by immunohistochemical staining for CD8; μCT image pre-processing (voxel resampling, image discretization and delineation of the volume of interest) and radiomic features (RFs) extraction (pyradiomics).

Results

We developed and optimized an efficient quantitative μCT imaging approach. Overall, 106 μCT RFs (shape, first- and second- order) were extracted and correlated with distinct TIME. Following redundant feature elimination (Spearman correlation, cut-off = 0.99) and Z-score standardization, we identified 12 μCT-RFs differentially regulated in T CD8 enriched vs depleted TIME (P < 0.05, Mann Whitney). The statistical performance of our CD8 signature, documented in both training and validation sets, was further implemented when pooled data were analysed (P < 0.01). Finally, by applying a reconstruction algorithm, we obtained a 3D map of CD8+ TILs, which enabled us to correlate the spatial distribution of immune cells with μCT textural parameters.

Conclusions

Our results document the feasibility and accuracy of radiomics to detect dramatic changes in T cells within the TIME, thus providing effective radio-immune signatures potentially translatable into clinical practice.

Legal entity responsible for the study

Institut Gustave Roussy.

Funding

ESMO (Translational Research Fellowship; Recipient: Dr. Giulia Mazzaschi).

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

10P - Cancer cells induce intracellular gap formation in sinusoidal endothelial cells to produce liver metastasis through pro-inflammatory paracrine mechanisms

Presentation Number
10P
Speakers
  • Hoang H. Truong (Hanoi, Viet Nam)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Engraftment of cancer cells to liver pre-metastatic niches requires complex interaction with liver sinusoidal endothelial cells (LSECs). Intracellular gap (iGap) formation in LSECs is caused by the destruction of fenestrae and appears under pathological conditions. Here, we aimed to explore the novel role of LSECs-iGap in liver metastasis of cancer cells.

Methods

Mouse models using acetaminophen (APAP) or thioacetamide (TAA) followed by intrasplenic injection of hepatocellular carcinoma (HCC) cell line, Hepa1-6 cells, to assess the LSECs-iGap formation. Functional effects of LSECs-iGap on cancer cells were analyzed using MMPs inducer, monocrotaline and inhibitor, doxycycline and MMP2/9 inhibitor. Morphological and molecular features of LSECs-iGap were examined using in vivo mouse models and in vitro co-culture system with electron microscopy, 3 dimensions tomographic reconstruction image (3D-TRI), RNA-sequencing, cytokine array and endothelial trans-endothelial migration assay. Biopsy specimens from 98 patients with HCC were statistically evaluated.

Results

APAP-induced liver injury and TAA-induced fibrotic liver resulted in LSECs-iGap formation, which positively correlated with increased numbers of metastatic foci after Hepa1-6 cells injection. In addition, Hepa1-6 cells induced IL-23-dependent TNF-α secretion by LSECs and triggered LSECs-iGap formation, toward which their processes protruded to transmigrate into the liver parenchyma. TNF-α caused depolymerization of F-actin and increased MMP9, ICAM1, and CXCLs expression in LSECs. Moreover, high expression of ICAM1 and MMP9 was significantly associated with intrahepatic metastasis and overall survival of patients with HCC. Blocking MMP9 activity by doxycycline or MMP-2/9 inhibitor eliminated LSECs-iGap formation and attenuated liver metastasis of Hepa1-6 cells.

Conclusions

This study revealed that cancer cells induced LSEC-iGap formation via pro-inflammatory paracrine mechanisms and proposed MMP9 as a novel target for blocking cancer cell metastasis to the liver.

Legal entity responsible for the study

H.H. Truong.

Funding

Japan Agency for Medical Research and Development.

Disclosure

All authors have declared no conflicts of interest.

Collapse
Basic science

11P - Targeting stromal cells to reverse immune suppression in triple-negative breast cancer

Presentation Number
11P
Speakers
  • Julia Chen (Darlinghurst, Australia)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Although there have been some positive results, the success of immune checkpoint inhibition in triple-negative breast cancer (TNBC) to date remains rather limited. A better understanding of mechanisms of immune evasion other than via PD1/PD-L1 axes and how to overcome these is therefore much needed to improve the efficacy of immunotherapy in this poor prognosis disease. Recent studies of the tumour microenvironment (TME) have elucidated the heterogeneity of stromal cells and suggested that stromal subsets play an important role in regulating anti-tumour immunity. Here we investigate how stromal cells contribute to the immunosuppressive TME and as a potential therapeutic target to overcome this.

Methods

Using co-culture models of patient-derived TNBC cancer-associated fibroblasts (CAFs) and peripheral blood mononuclear cells (PBMCs) from healthy donors, we applied flow cytometry and single-cell RNA sequencing (scRNAseq) to assess stromal-immune interactions in vitro. A high throughput drug screen was performed to identify novel stromal-targeting drug candidates which could reverse stromal-induced immunosuppression in our coculture models.

Results

TNBC CAFs suppress the proliferative capability of both CD4 and CD8 T cells in vitro. scRNAseq of these co-culture models demonstrate that CAF-cocultured T cells are driven into LAG3+ exhausted state enriched for canonical pathways of immunosuppressive cytokine signalling. Drug screen identified Talabostat as a novel candidate which shifts stromal subtype composition from aSMA-positive to aSMA-negative phenotypes. Talabostat treatment in the co-culture models reverses CAF-induced suppression of T cell proliferation.

Conclusions

Talabostat reverses stromal-mediated T cell suppression and is a potential therapeutic strategy to elicit a more effective immune response.

Legal entity responsible for the study

The authors.

Funding

US Department of Defense grant.

Disclosure

E. Lim: Financial Interests, Institutional, Advisory Board: Gilead, Novartis, Pfizer, Roche, AstraZeneca, MSD, Lilly; Financial Interests, Institutional, Invited Speaker: Roche, Gilead, Novartis, Lilly, AstraZeneca; Financial Interests, Institutional, Research Grant: Pfizer, Novartis; Financial Interests, Institutional, Steering Committee Member: Roche, Lilly, AstraZeneca; Financial Interests, Institutional, Local PI: Novartis, Gilead; Non-Financial Interests, Leadership Role, Scientific Advisory Committee: Breast Cancer Trials Australia; Non-Financial Interests, Leadership Role, Principal Cancer Theme Lead, Faculty of Medicine: University of New South Wales; Non-Financial Interests, Leadership Role, Faculty: Garvan Institute of Medical Research; Non-Financial Interests, Leadership Role, Director Cancer Research: St Vincent's Hospital Sydeny. A. Swarbrick: Financial Interests, Personal and Institutional, Financially Compensated Role, Paid Consultant: Phenomic AI. All other authors have declared no conflicts of interest.

Collapse
Basic science

12P - Immuno-suppressive role of tumour-derived GDF-15 on myeloid cells

Presentation Number
12P
Speakers
  • Christine Schuberth-Wagner (Planegg-Martinsried, Germany)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Growth and differentiation factor 15 (GDF-15), a divergent member of the TGF-β superfamily, shows high expression during pregnancy. Beyond that, GDF-15 is induced in stressed and damaged tissues limiting inflammation. GDF-15 also functions as a prognostic marker in numerous cancers and was shown to be a key inhibitor of T-cell infiltration in solid tumors. However, the impact of GDF-15 on other immune cells in the tumor microenvironment (TME) is still poorly understood.

Methods

M1 and M2 macrophages were derived from either the human monocytic THP-1 cell line or monocytes from healthy donors by incubation with IFNγ and LPS (M1) or IL-4 and IL-13 (M2) in combination with or without GDF-15. Analysis was performed by Western Blot, flow cytometry and RT-qPCR. To investigate the role of GDF-15 in vivo we inoculated NOG mice subcutaneously with SK-Mel5 GDF15 -/- cells. On day 35/36 tumors were assessed on myeloid immune cell subpopulations by flow cytometry.

Results

Our initial studies have shown that monocytes are sensitive to GDF-15 mediated inhibition of tumor infiltration. Now we wanted to understand how GDF-15 affects the myeloid compartment beyond infiltration. In primary cells, GDF-15 exposure of monocytes during IFNγ or LPS induced M1 polarization led to a reduction of activation marker expression such as HLA-DR, CD80 and CD86 and an altered cytokine secretion profile. M2 macrophages did not show differences in their activation pattern. This effect was transferable to THP-1 derived M1 macrophages. TGF-ß was not able to exert this biologic effect. Activation was restored by addition of a GDF-15 neutralizing antibody. In vivo, GDF-15 had no direct effect on tumor growth, yet we observed that loss of GDF-15 altered the TME and led to an increase of activated myeloid cells such as M1 macrophages and dendritic cells.

Conclusions

GDF-15 secretion helps tumors to generate a TME that is poorly infiltrated by anti-tumoral immune cells. GDF-15 counteracts myeloid activation in the TME, a hallmark of initiation of antitumoral immune responses. By inducing a more pro-inflammatory tumor phenotype, anti-GDF-15 antibodies may synergize with other immunotherapeutic agents. A clinical phase 2 trial combining anti-GDF-15 (CTL-002) with anti-PD-1 (NCT04725474) is ongoing.

Legal entity responsible for the study

CatalYm GmbH.

Funding

CatalYm GmbH.

Disclosure

C. Schuberth-Wagner: Financial Interests, Personal, Full or part-time Employment: CatalYm GmbH; Financial Interests, Personal, Stocks/Shares: CatalYm GmbH, Merck & Co; Financial Interests, Personal, Royalties: Rigontec GmbH; Non-Financial Interests, Advisory Role: RNHale GmbH. I. Giese, M. Kist, N. Vashist, S. Genßler, M. Auer, K. Klar: Financial Interests, Personal, Full or part-time Employment: CatalYm GmbH. J. Weigandt: Financial Interests, Personal, Full or part-time Employment, Scientist Role: Catalym; Financial Interests, Personal, Stocks/Shares, small number of shares held privately: AstraZeneca, Moderna. J. Wischhusen: Financial Interests, Personal, Advisory Board: CatalYm; Financial Interests, Personal, Stocks/Shares: CatalYm; Financial Interests, Personal, Advisory Board, Fees received via University of Würzburg as compensation for inventorship: CatalYm; Financial Interests, Institutional, Funding: CatalYm. E. Leo: Financial Interests, Personal, Advisory Board, Advisory role: T-knife GmbH; Financial Interests, Personal, Full or part-time Employment, CMO: CatalYm GmbH; Financial Interests, Personal, Stocks/Shares, Share options: CatalYm GmbH, T-knife GmbH. M. Haake: Financial Interests, Personal, Full or part-time Employment: CatalYm GmbH; Financial Interests, Personal, Stocks/Shares, Co-Founder: CatalYm GmbH; Financial Interests, Personal, Advisory Board: CatalYm GmbH; Non-Financial Interests, Other, Co-founder: CatalYm GmbH. All other authors have declared no conflicts of interest.

Collapse
Basic science

13P - Disrupting the immunosuppressive tumor microenvironment using genetically engineered macrophages for triple-negative breast cancer therapy

Presentation Number
13P
Speakers
  • Sabrina Traxel (Zürich, Switzerland)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer and accounts for 40% of breast cancer deaths. Currently, surgery and chemotherapy are standard therapy in TNBC, whereas immunotherapies often fail due to a cold tumor microenvironment (TME), which is mainly generated by tumor associated macrophages (TAMs). Importantly, immunosuppressive M2-like TAMs are associated with worse outcome in TNBC patients. Cytokines like IL-10 and TGFβ induce M2-like TAMs, which inhibit the adaptive immune response and promote tumor growth, migration, and vascularization. In contrast, M1-like macrophages induce an anti-tumor immune response and tumor cell death. Thus, we aim to develop an adoptive cell therapy employing macrophages that are polarized towards a M1-like phenotype by the immunosuppressive cytokines, which will generate an inflamed TME.

Methods

To achieve M1 polarization in a M2-inducing TME, we genetically modified macrophages to express a chimeric cytokine receptor (ChCR), which will induce an M1 signaling upon stimulation with M2 inducing cytokines. Here, we evaluated two ChCR variants activated by IL-10 and TGFβ, respectively. We transduced monocytes to express the ChCR using lentiviruses. Upon differentiation to macrophages, we assessed M1 polarization by analyzing surface marker expression and the secretome. Moreover, we assessed their effect on TNBC cell line viability.

Results

We could successfully transduce macrophages at 80% and confirmed ChCR expression and M1 signaling. Furthermore, we could show that stimulation of ChCR macrophages by IL-10 or TGFβ switches them towards an M1-like phenotype, as analyzed by surface marker expression and IP-10 secretion. Moreover, we found that the viability of TNBC cell lines was reduced upon treatment with supernatants of TGFβ or IL-10 stimulated ChCR macrophages.

Conclusions

We could show that we can engineer macrophages to switch towards an M1-like phenotype in an M2-inducing environment. Thus, ChCR macrophages represent a promising strategy to disrupt the immunosuppressive TME. Currently, we are assessing the effect of ChCR macrophages on immune activation in vitro and their phenotype in TNBC co-cultures.

Legal entity responsible for the study

The authors.

Funding

Gebauer Stiftung, Lotte und Adolf Hotz-Sprenger-Stiftung, Claudia von Schilling, Foundation for Breast Cancer Research, OPO-Stiftung, Stiftung zur Krebsbekämpfung, Alfred und Anneliese Sutter-Stöttner Stiftung, Dr. Arnold U. und Susanne, Huggenberger-Bischoff Stiftung, Novartis Foundation for Medical-Biological Research, UZH Bio & MedTech Entrepreneur Fellowship, UZH Postdoc Grant.

Disclosure

R.F. Speck: Other, Personal and Institutional, Other, Inventor of pending patent EP 22/215498 related to project: University of Zurich. S. Bredl: Financial Interests, Personal and Institutional, Other, Inventor of pending patent EP 22/215498 related to project: University of Zurich. All other authors have declared no conflicts of interest.

Collapse
Basic science

14P - Peripheral immune kinetics in survival prediction of small cell lung cancer patients treated with immune checkpoint blockade therapy

Presentation Number
14P
Speakers
  • MIGUEL A. GALINDO CAMPOS (Barcelona, Spain)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

Small cell lung cancer (SCLC) is a lethal neoplasia. Chemotherapy (Ct) plus immune checkpoint blockade therapy (ICBt) is now the standard of care although limited survival benefit. Lack of biomarkers of response leads to suboptimal patient selection, distorting results of clinical trials. Deciphering the ICBt-driven peripheral immune events may recognize those patients who benefit the most. Here, we characterize early peripheral immune kinetics in SCLC patients treated with Ct + ICBt with improved survival.

Methods

SCLC patients from 8 centers were prospectively recruited into 3 cohorts. Cohort 1 include patients treated with Ct (n=24). Cohort 2 (n=37) comprise patients treated with Ct + anti-CTLA-4, and cohort 3 (n=20) patients treated with Ct + anti-PD-1/PD-L1. Peripheral blood mononuclear cells were obtained prior to treatment and after the first dose of ICBt. Variation in proliferation, senescence, adhesion, immunosuppression, and checkpoints markers was assessed by FACS. Kaplan–Meier and log-rank test were used for survival analysis. MaxStat was used to calculate cut points. A p-value <0.05 was considered statistically significant.

Results

We found 6 independent cellular subsets which modulation right after the start of ICBt identify patients with improved survival. An increase in CD8+CD103+Ki67+ cells identify survival benefit in cohorts 1 and 3 (p=0.043; 0.0033). An upregulation of Ki67 in total CD4+ (p=0.012; 0.0027) and CD4+PD-1+ T cells (p=0.026; 0.027), predicts long survival in ICBt cohorts 2 and 3, while a downregulation of CD4+ICOS+ T cells (p=0.025; 0.011) identified survival benefit in these ICBt cohorts. Expansion of Ki67+ and ICOS+ (p=0.0024; 0.0074) CD8+ T cells was also observed in CD8+ T cells from long survivors exclusively from cohort 2.

Conclusions

Expansion of CD8+CD103+Ki67+, CD4+Ki67+, and CD4+PD-1+Ki67+, or a reduction in CD4+ICOS+ circulating T cells subsets identify long survival patients treated with anti-CTLA-4 or anti-PD-1/PD-L1, while an increase in CD8+Ki67+ or CD8+ICOS+ cells predicts benefit only in patients treated with anti-CTLA-4. Assessment of the immune kinetics of these peripheral cellular subsets identify patients who benefit the most from ICBt in SCLC.

Legal entity responsible for the study

The authors.

Funding

Fundació Amics de l'Hospital del Mar.

Disclosure

E. Arriola: Financial Interests, Personal and Institutional, Advisory Role, Speaker, Institutional funding, Travel and educational expenses: BMS, MSD; Financial Interests, Personal and Institutional, Advisory Role, Speaker, Institutional and Research funding, Travel and educational expenses: Roche; Financial Interests, Personal and Institutional, Advisory Role, Speaker, Institutional and Research funding: Pfizer; Financial Interests, Personal, Advisory Role, Speaker, Travel and educational expenses: Lilly; Financial Interests, Personal and Institutional, Advisory Role, Speaker, Institutional funding: AstraZeneca; Financial Interests, Personal, Advisory Role, Speaker: Boehringer Ingelheim, Takeda; Other, Personal, Member of Board of Directors, Co-founder: TrialingHealth. All other authors have declared no conflicts of interest.

Collapse
Basic science

15P - CBL E3 ubiquitin ligases are key inhibitory regulators in PD-1/LAG-3 co-signaling in human cancers, targeted through bispecific co-blockade

Presentation Number
15P
Speakers
  • Luisa Chocarro (Pamplona, Spain)
Onsite Poster display date
Sunday, 22 October 2023

Abstract

Background

A significant number of cancer patients do not benefit from PD-L1/PD-1 blockade immunotherapies. PD-1 and LAG-3 co-upregulation in T-cells is one of the major mechanisms of resistance by establishing a highly dysfunctional state in T-cells.

Methods

A high-throughput screening was performed of PD-1/LAG-3 multiomic expression profiles in public databases of human cancer biopsies, to establish relationships between infiltrating tumor-infiltrating PD-1/LAG-3 T cells with biomarkers both in T cells and within the tumour microenvironment. These results were validated in engineered T-cell lines with constitutively active PD-1, LAG-3 pathways, and their combination, analysed by high-throughput quantitative proteomics and validated by conventional molecular techniques on primary T cells from NSCLC patients.

Results

The high-throughput multiomic human cancers screening and the experimental proteomic T cell lines uncovered a strong PD-1/LAG-3 dysfunctionality signature that was found which regulated immune, proteomic, metabolic, genetic, and epigenetic pathways. These mainly relied on differential regulation of E3 ubiquitin ligases CBL-B and C-CBL. Notably, LAG-3 expression has never been associated to CBL ubiquitin ligases before.

Conclusions

PD-1/LAG-3 co-signaling profile uncovers a highly dysfunctional regulated programme. Co-blockade with a bispecific drug but not with a combination of anti-PD-1/anti-LAG-3 antibodies achieved both CBL-B and C-CBL inhibition, reverting T-cell dysfunctionality in lung cancer patients resistant to PD-L1/PD- 1 blockade. These results will help identifying the mechanisms of intrinsic resistance to PD-1 blockade mediated by LAG-3 co-signaling.

Legal entity responsible for the study

Navarrabiomed.

Funding

This research was supported by: The Spanish Association against Cancer (AECC), PROYE16001ESCO; Instituto de Salud Carlos III (ISCIII)-FEDER Project grants FIS PI20/00010, COV20/00000, and TRANSPOCART ICI19/00069; Biomedicine Project Grant from the Department of Health of the Government of Navarre-FEDER funds (BMED 050-2019, 51-2021); Strategic projects from the Department of Industry, Government of Navarre (AGATA, Ref. 0011-1411-2020-000013; LINTERNA, Ref. 0011-1411-2020-000033; DESCARTHES, 0011-1411-2019-000058); European Union Horizon 2020 ISOLDA project, under grant agreement ID: 848166. L.C. is financed by Instituto de Salud Carlos III (ISCIII), co-financed by FEDER funds, "Contratos PFIS: contratos predoctorales de formación en investigación en salud" (FI21/00080); M.E. is financed by the Navarrabiomed- Fundación Miguel Servet predoctoral contract.

Disclosure

C.J. Edwards: Other, Personal, Other, Declares to be inventor of the Humabody CB213 (WO/2019/158942. 603 Crescendo Biologics Ltd.): Crescendo Biologics. J. Leggs: Other, Declares to be e inventor of the Humabody CB213 (WO/2019/158942. 603 Crescendo Biologics Ltd.): Crescendo Biologics. D. Escors: Other, Declares to be e inventor of the Humabody CB213 (WO/2019/158942. 603 Crescendo Biologics Ltd.): Navarrabiomed. All other authors have declared no conflicts of interest.

Collapse