scholarly journals Cancer Cell–Intrinsic Expression of MHC Class II Regulates the Immune Microenvironment and Response to Anti–PD-1 Therapy in Lung Adenocarcinoma

2020 ◽  
Vol 204 (8) ◽  
pp. 2295-2307 ◽  
Author(s):  
Amber M. Johnson ◽  
Bonnie L. Bullock ◽  
Alexander J. Neuwelt ◽  
Joanna M. Poczobutt ◽  
Rachael E. Kaspar ◽  
...  
2020 ◽  
Author(s):  
Amber M. Johnson ◽  
Bonnie L. Bullock ◽  
Alexander J. Neuwelt ◽  
Joanna M. Poczobutt ◽  
Rachael E. Kaspar ◽  
...  

AbstractMHC class II (MHCII) expression is usually restricted to antigen presenting cells, but can be expressed by cancer cells. We examined the effect of cancer cell-intrinsic MHC class II (csMHCII) expression in lung adenocarcinoma on T cell recruitment to tumors and response to anti-PD-1 therapy. The functional significance of altering csMHCII expression was explored using two orthotopic immunocompetent murine models of non-small cell lung cancer: CMT167 (CMT) and Lewis Lung Carcinoma (LLC). We previously showed that CMT167 tumors are eradicated by anti-PD1 therapy, while LLC tumors are resistant. RNA-seq analysis of cancer cells recovered from tumors revealed that csMHCII correlated with response to anti-PD1 therapy, with immunotherapy-sensitive CMT167 cells being csMHCII positive, while resistant LLC cells were csMHCII negative. To test the functional effects of csMHCII, MHCII expression was altered on the cancer cells through loss- and gain-of-function of CIITA, a master regulator of the MHCII pathway. Loss of CIITA in CMT167 decreased csMHCII, and converted tumors from anti-PD-1-sensitive to anti-PD-1-resistant. This was associated with decreased T cell infiltration, lower levels of Th1 cytokines, increased B cell number and decreased macrophage recruitment. Conversely, overexpression of CIITA in LLC cells resulted in csMHCII in vitro and in vivo. Enforced expression of CIITA increased T cell infiltration and sensitized tumors to anti-PD-1 therapy. csMHCII expression was also examined in a subset of surgically resected human lung adenocarcinomas by multispectral imaging, provided a survival benefit and positively correlated with T cell infiltration. These studies demonstrate a functional role for csMHCII in regulating T cell infiltration and sensitivity to anti-PD-1.


Author(s):  
Zhiwen Luo ◽  
Xiao Chen ◽  
Yefan Zhang ◽  
Zhen Huang ◽  
Hong Zhao ◽  
...  

Background: Post-surgical recurrence of the metastatic colorectal cancer (mCRC) remains a challenge, even with adjuvant therapy. Moreover, patients show variable outcomes. Here, we set to identify gene models based on the perspectives of intrinsic cell activities and extrinsic immune microenvironment to predict the recurrence of mCRC and guide the adjuvant therapy.Methods: An RNA-based gene expression analysis of CRC samples (total = 998, including mCRCs = 344, non-mCRCs = 654) was performed. A metastasis-evaluation model (MEM) for mCRCs was developed using the Cox survival model based on the prognostic differentially expressed genes between mCRCs and non-mCRCs. This model separated the mCRC samples into high- and low-recurrence risk clusters that were tested using machine learning to predict recurrence. Further, an immune prognostic model (IPM) was built using the COX survival model with the prognostic differentially expressed immune-related genes between the two MEM risk clusters. The ability of MEM and IPM to predict prognosis was analyzed and validated. Moreover, the IPM was utilized to evaluate its relationship with the immune microenvironment and response to immuno-/chemotherapy. Finally, the dysregulation cause of IPM three genes was analyzed in bioinformatics.Results: A high post-operative recurrence risk was observed owing to the downregulation of the immune response, which was influenced by MEM genes (BAMBI, F13A1, LCN2) and their related IPM genes (SLIT2, CDKN2A, CLU). The MEM and IPM were developed and validated through mCRC samples to differentiate between low- and high-recurrence risk in a real-world cohort. The functional enrichment analysis suggested pathways related to immune response and immune system diseases as the major functional pathways related to the IPM genes. The IPM high-risk group (IPM-high) showed higher fractions of regulatory T cells (Tregs) and smaller fractions of resting memory CD4+ T cells than the IPM-low group. Moreover, the stroma and immune cells in the IPM-high samples were scant. Further, the IPM-high group showed downregulation of MHC class II molecules. Additionally, the Tumor Immune Dysfunction and Exclusion (TIDE) algorithm and GDSC analysis suggested the IPM-low as a promising responder to anti-CTLA-4 therapy and the common FDA-targeted drugs, while the IPM-high was non-responsive to these treatments. However, treatment using anti-CDKN2A agents, along with the activation of major histocompatibility complex (MHC) class-II response might sensitize this refractory mCRC subgroup. The dysfunction of MEIS1 might be the reason for the dysregulation of IPM genes.Conclusions: The IPM could identify subgroups of mCRC with a distinct risk of recurrence and stratify the patients sensitive to immuno-/chemotherapy. Further, for the first time, our study highlights the importance of MHC class-II molecules in the treatment of mCRCs using immunotherapy.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 4043-4043
Author(s):  
Zhiwen Luo ◽  
Xinyu Bi

4043 Background: Recurrence of concurrent metastatic colorectal cancers (mCRCs) after surgery is still a challenge. But mCRCs’ outcomes are heterogeneous, and no clinicopathological methods can predict its recurrence and guide postoperative treatment from an intrinsic cell activities and extrinsic immune microenvironment perspective. We aimed to identify such gene models. Methods: Gene expression analysis on CRCs. Based on metastasis-related genes, a metastatic evaluation model (MEM) was developed, dividing mCRCs into high and low recurrence risk clusters. Machine learning tested MEM’s importance to predict recurrence. Further investigating MEM’s two clusters made an immune prognostic model (IPM) with immune genes differentially expressed between MEM clusters. The predictive performance of MEM and IPM on prognosis was comprehensively analyzed and validated. The mechanism of IPM on the immune microenvironment and response to immuno/chemotherapy was analyzed extensively. Results: RNA data of 998 CRCs were analyzed. High postoperative recurrence risk in mCRCs was owing to immune response’s down-regulation, which was influenced by 3 MEM genes ( BAMBI, F13A1, LCN2) and their related 3 IPM genes ( SLIT2, CDKN2A, CLU). MEM and IPM were developed and validated on 239 mCRCs to differentiate a low and high recurrence risk (AUCs > 0.7). Functional enrichment analysis showed immune response and immune system diseases pathway represented the major function and pathway related to IPM gene. IPM high-risk group (IPM-high) had higher fractions of Tregs ( P= 0.04), lower fractions of resisting memory CD4+ T cells ( P= 0.02) than IPM-low. And stroma and immune cells in IPM-high samples were scant ( P= 0.0002, 0.001, respectively). In IPM-high, MHC class II molecules all down-expressed, and DNA methylation disordered. TIDE algorithm and GDSC analysis discovered IPM-low was more promising to respond to both anti-CTLA4 therapy ( P= 0.005) and common FDA targeted drugs ( P< 0.05), while IPM-high had nonresponse to both of them. But anti-CDKN2A agent with activation of MHC class II response might reverse the dilemma of this refractory mCRCs subgroup. Conclusions: Postoperative recurrence of mCRC is strongly related to immune microenvironment. Our two relative gene models could identify subgroups of mCRC with different recurrence risk, and stratify mCRCs sensitive to immune/chemotherapy, even highlight the ignored importance of MHC class II molecules on immunotherapy in mCRCs for the first time.


2020 ◽  
Author(s):  
Amber M. Johnson ◽  
Bonnie L. Bulock ◽  
Alexander J. Neuwelt ◽  
Erin L. Schenk ◽  
Eric T. Clambey ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document