151
|
Goc J, Sonnenberg GF. Harnessing Microbiota to Improve Immunotherapy for Gastrointestinal Cancers. Cancer Immunol Res 2022; 10:1292-1298. [PMID: 36166399 PMCID: PMC10424780 DOI: 10.1158/2326-6066.cir-22-0164] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 06/20/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022]
Abstract
Immune checkpoint blockade has revolutionized opportunities for therapeutic intervention in cancer but demonstrates a low frequency of response in most patients and in some common types of tumors. An emerging paradigm supports the notion that trillions of normally beneficial microbes inhabiting the gastrointestinal tract, termed the microbiota, critically impact the success or failure of antitumor immunity induced by immune checkpoint blockade. Here, we briefly summarize the current knowledge on how interactions between the microbiota and immune system are contributing to the outcome of cancer immunotherapy. We propose that this immune-microbiota dialogue is particularly important in gastrointestinal cancers that exhibit striking resistance to immune checkpoint blockade and inherently develop in a unique environment that is rich in both immune-cell networks and direct exposure to the microbiota. Finally, we focus on how future studies should determine whether microbiota can be harnessed as a strategy to boost antitumor immunity in these contexts and beyond. See related article, p. 1291.
Collapse
Affiliation(s)
- Jeremy Goc
- Division of Gastroenterology & Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, New York
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
| | - Gregory F Sonnenberg
- Division of Gastroenterology & Hepatology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, New York
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, New York
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, New York
| |
Collapse
|
152
|
Taieb J, Svrcek M, Cohen R, Basile D, Tougeron D, Phelip JM. Deficient mismatch repair/microsatellite unstable colorectal cancer: Diagnosis, prognosis and treatment. Eur J Cancer 2022; 175:136-157. [PMID: 36115290 DOI: 10.1016/j.ejca.2022.07.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/08/2022] [Accepted: 07/17/2022] [Indexed: 11/03/2022]
Abstract
Microsatellite unstable (MSI) colorectal cancers (CRCs) are due to DNA mismatch repair (MMR) deficiency and occurs in15% of non-metastatic diseases and 5% in the metastatic setting. Nearly 30% of MSI CRCs occur in a context of constitutional mutation of the MMR system (Lynch syndrome). Others are sporadic cancers linked to a hypermethylation of the MLH-1 promoter. The pathogenic alterations of MMR genes lead to the accumulation of frequent somatic mutational events and these tumours arbour a high antigen burden and are highly infiltrated with cytotoxic T-cell lymphocytes. Microsatellite instability/DNA mismatch repair deficiency (MSI/dMMR) status has prognostic and predictive implications in non-metastatic and metastatic CRCs. The prognostic value of MSI status in non-metastatic CRCs has been studied extensively, yet the data are more limited for its predictive value in terms of adjuvant chemotherapy efficacy. In both cases (metastatic and non-metastatic settings) treatment with immune check-point inhibitors (ICIs) have shown a remarkable effectiveness in the context of MSI/dMMR status. Indeed, recent data from prospective cohorts and randomised trials have shown a dramatical improvement of survival with immunotherapy (programmed death-ligand 1 [PD-(L)1] cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] blockage) in metastatic or non-metastatic MSI/dMMR CRC. In this review we report and discuss how and for whom to test for the MSI/dMMR phenotype, as well as the prognostic value of this phenotype and the new treatment recommendations options for this unique CRC population. Despite their efficacy, primary and secondary resistance to immune checkpoint inhibitors (ICIs) are observed in more than 50% MSI-H/dMMR CRC patients and in the future how to identify these patients and to overcome resistance will be an important challenge.
Collapse
Affiliation(s)
- Julien Taieb
- Université Paris-Cité, Department of Gastroenterology and Digestive Oncology, Georges Pompidou European Hospital, SIRIC CARPEM, Paris, France.
| | - Magali Svrcek
- Sorbonne Université, Department of Pathology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer et SIRIC CURAMUS, Centre de Recherche Saint Antoine, Paris, France
| | - Romain Cohen
- Sorbonne Université, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP, and INSERM UMRS 938, Équipe Instabilité des Microsatellites et Cancer, Équipe Labellisée par la Ligue Nationale Contre le Cancer et SIRIC CURAMUS, Centre de Recherche Saint Antoine, Paris, France
| | - Debora Basile
- Department of Medical Oncology, San Giovanni di Dio Hospital, 88900 Crotone, Italy
| | - David Tougeron
- Université de Poitiers, Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Jean-Marc Phelip
- University Hospital of Saint Etienne, Saint Etienne, France; Unité HESPER EA-7425 Université Jean Monnet/Claude Bernard Lyon 1, France
| |
Collapse
|
153
|
Geng Y, Feng J, Huang H, Wang Y, Yi X, Wei S, Zhang M, Li Z, Wang W, Hu W. Single-cell transcriptome analysis of tumor immune microenvironment characteristics in colorectal cancer liver metastasis. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1170. [PMID: 36467341 PMCID: PMC9708492 DOI: 10.21037/atm-22-5270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 11/03/2022] [Indexed: 10/22/2023]
Abstract
BACKGROUND Liver metastasis is the leading cause of death in colorectal cancer (CRC) patients, and the precise mechanisms remain unclear. In this study, single-cell RNA sequencing (scRNA-seq) was used to analyze the cellular and molecular heterogeneity between CRC primary lesion and corresponding liver metastasis, and to clarify the characteristics of the tumor microenvironment (TME) in synchronous liver metastasis of CRC. METHODS A case of microsatellite stable (MSS) sigmoid carcinoma with synchronous liver metastasis was selected, and tissues from the primary tumor and the liver metastasis were collected for scRNA-seq. The EdgeR package software was used to identify the differentially expressed genes between cells. Gene Set Enrichment Analysis (GSEA) was performed and the clusterProfiler R package was used for Gene Ontology (GO) enrichment analysis. The SCENIC and CellphoneDB packages were used to reconstruct the transcriptional regulatory networks and to analyze the intercellular interaction network, respectively. RESULTS Compared to the primary tumor, the proportion of myeloid cells in the metastatic tumor was significantly increased, while B cells and plasma cells were decreased. In the metastatic tumor, the myeloid-derived suppressor cell (MDSC) characteristic gene, mannose receptor C-type 1 (MRC1) and tumor associated macrophage 2 (TAM2)-related gene, were highly expressed. Furthermore, angiogenesis, oxidative phosphorylation, and endothelial mesenchymal transition (EMT) of myeloid cells were also significantly enhanced. There were less myeloid cells in primary tumors, and these were mainly monocytes and TAM1; while the number of TAM2 was significantly upregulated in the metastatic samples. In liver metastasis, the T cell population was exhausted, and this was accompanied by a significant increase in the number of CD4+ T cells and a decrease in the number of CD8+ T cells. Furthermore, some immune checkpoint molecules were highly expressed. Interactions between myeloid cells and other cell populations appeared to be strong. CONCLUSIONS The TME of CRC liver metastasis is significantly immunosuppressed. Interactions between myeloid cells and other cell populations in the TME contribute to the establishment of a pro-metastatic niche that promotes colonization and growth of CRC cells in the liver. TAMs may be a potential immunotherapeutic target for MSS CRC.
Collapse
Affiliation(s)
- Yiting Geng
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Feng
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ying Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xing Yi
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Shanshan Wei
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Mingyue Zhang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhong Li
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wei Wang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenwei Hu
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
154
|
Lote H, Starling N, Pihlak R, Gerlinger M. Advances in immunotherapy for MMR proficient colorectal cancer. Cancer Treat Rev 2022; 111:102480. [DOI: 10.1016/j.ctrv.2022.102480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/23/2022] [Accepted: 10/26/2022] [Indexed: 11/02/2022]
|
155
|
Shigaki T, Fujiyoshi K, Sudo T, Kawahara A, Nakane H, Yomoda T, Nagasu S, Kinugasa T, Akiba J, Fujita F, Akagi Y. Mismatch repair proteins expression and tumor-infiltrating T-cells in colorectal cancer. Oncol Lett 2022; 24:396. [PMID: 36276483 PMCID: PMC9533364 DOI: 10.3892/ol.2022.13516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/31/2022] [Indexed: 11/06/2022] Open
Abstract
Microsatellite instability (MSI) and tumor mutational burden (TMB) are indicators of the tumor mutational load, which can lead to immune cell recruitment. By contrast, the number of tumor-infiltrating T cells (TITs) is indicative of the host immune response to tumor cells. The present study evaluated if the expression of mismatch repair (MMR) proteins can be used as a precise tool to assess immunogenicity in the tumor microenvironment. A total of 73 colorectal cancer cases were enrolled in the present study. MMR protein expression was assessed using four-antibodies immunohistochemistry (IHC) targeting MLH1, MSH2, MSH6 and PMS2. TIT was assessed through IHC by counting CD3+ and CD8+ cells in tumor. The enrolled cases were classified into four groups according to MMR and TIT status i) Mismatch repair-proficient (pMMR) and a high number of TITs (pMMR/TIT-H); ii) pMMR and a low number of TITs (pMMR/TIT-L); iii) mismatch repair-deficient (dMMR) and TIT-H (dMMR/TIT-H); and iv) dMMR/TIT-L]. The present study evaluated the clinicopathological characteristics of the four groups, in addition to the difference of TMB. TMB analysis was counted the number of the somatic mutations through multi-genes panel using next-generation sequencing. Clinicopathological characteristics, including age, sex, pathological depth of invasion and lymph node metastasis, were not found to be statistically different between dMMR/TIT-H and dMMR/TIT-L groups. Tumors among pMMR/TIT-H group were associated with poorly differentiation compared with those in pMMR/TIT-L group (P=0.025). The median TMB among the dMMR/TIT-H group was the highest in four groups but the median TMB was <10 muts/Mb in dMMR/TIT-L, pMMR/TIT-H and pMMR/TIT-L groups, respectively. However, one tumor in the pMMR/TIT-H group showed high TMB. The present findings suggest that assessing MMR status alone may not be sufficient to precisely evaluate the antitumor immune response in the tumor microenvironment.
Collapse
Affiliation(s)
- Takahiro Shigaki
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Kenji Fujiyoshi
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Tomoya Sudo
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Akihiro Kawahara
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Hiroyuki Nakane
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Takato Yomoda
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Sachiko Nagasu
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Tetsushi Kinugasa
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Jun Akiba
- Department of Pathology, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Fumihiko Fujita
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| | - Yoshito Akagi
- Department of Surgery, Kurume University School of Medicine, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
156
|
Zhang X, Qin X, Yu T, Wang K, Chen Y, Xing Q. Chromatin regulators-related lncRNA signature predicting the prognosis of kidney renal clear cell carcinoma and its relationship with immune microenvironment: A study based on bioinformatics and experimental validation. Front Genet 2022; 13:974726. [PMID: 36338996 PMCID: PMC9630733 DOI: 10.3389/fgene.2022.974726] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Kidney Renal Clear cell carcinoma (KIRC) is a major concern in the urinary system. A lot of researches were focused on Chromatin Regulators (CRs) in tumors. In this study, CRs-related lncRNAs (CRlncRNAs) were investigated for their potential impact on the prognosis of KIRC and the immune microenvironment. Methods: The TCGA database was used to obtain transcriptome and related clinical information. CRs were obtained from previous studies, whereas CRlncRNAs were obtained by differential and correlation analysis. We screened the lncRNAs for the signature construction using regression analysis and LASSO regression analysis. The effectiveness of the signature was evaluated using the Kaplan-Meier (K-M) curve and Receiver Operating Characteristic curve (ROC). Additionally, we examined the associations between the signature and Tumor Microenvironment (TME), and the efficacy of drug therapy. Finally, we further verified whether these lncRNAs could affect the biological function of KIRC cells by functional experiments such as CCK8 and transwell assay. Results: A signature consisting of 8 CRlncRNAs was constructed to predict the prognosis of KIRC. Quantitative Real-Time PCR verified the expression of 8 lncRNAs at the cell line and tissue level. The signature was found to be an independent prognostic indicator for KIRC in regression analysis. This signature was found to predict Overall Survival (OS) better for patients in the subgroups of age, gender, grade, stage, M, N0, and T. Furthermore, a significant correlation was found between riskScore and immune cell infiltration and immune checkpoint. Finally, we discovered several drugs with different IC50 values in different risk groups using drug sensitivity analysis. And functional experiments showed that Z97200.1 could affect the proliferation, migration and invasion of KIRC cells. Conclusion: Overall, the signature comprised of these 8 lncRNAs were reliable prognostic biomarkers for KIRC. Moreover, the signature had significant potential for assessing the immunological landscape of tumors and providing individualized treatment.
Collapse
Affiliation(s)
- Xinyu Zhang
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xinyue Qin
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong University, Nantong, Jiangsu, China
| | - Tiannan Yu
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Kexin Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yinhao Chen
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- *Correspondence: Qianwei Xing, ; Yinhao Chen,
| | - Qianwei Xing
- Department of Urology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
- *Correspondence: Qianwei Xing, ; Yinhao Chen,
| |
Collapse
|
157
|
Huang EM, Ma N, Ma T, Zhou JY, Yang WS, Liu CX, Hou ZH, Chen S, Zong Z, Zeng B, Li YR, Zhou TC. Cuproptosis-related long non-coding RNAs model that effectively predicts prognosis in hepatocellular carcinoma. World J Gastrointest Oncol 2022; 14:1981-2003. [PMID: 36310708 PMCID: PMC9611437 DOI: 10.4251/wjgo.v14.i10.1981] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/29/2022] [Accepted: 08/17/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cuproptosis has recently been considered a novel form of programmed cell death. To date, long-chain non-coding RNAs (lncRNAs) crucial to the regulation of this process remain unelucidated.
AIM To identify lncRNAs linked to cuproptosis in order to estimate patients' prognoses for hepatocellular carcinoma (HCC).
METHODS Using RNA sequence data from The Cancer Genome Atlas Live Hepatocellular Carcinoma (TCGA-LIHC), a co-expression network of cuproptosis-related genes and lncRNAs was constructed. For HCC prognosis, we developed a cuproptosis-related lncRNA signature (CupRLSig) using univariate Cox, lasso, and multivariate Cox regression analyses. Kaplan-Meier analysis was used to compare overall survival among high- and low-risk groups stratified by median CupRLSig risk score. Furthermore, comparisons of functional annotation, immune infiltration, somatic mutation, tumor mutation burden (TMB), and pharmacologic options were made between high- and low-risk groups.
RESULTS Three hundred and forty-three patients with complete follow-up data were recruited in the analysis. Pearson correlation analysis identified 157 cuproptosis-related lncRNAs related to 14 cuproptosis genes. Next, we divided the TCGA-LIHC sample into a training set and a validation set. In univariate Cox regression analysis, 27 LncRNAs with prognostic value were identified in the training set. After lasso regression, the multivariate Cox regression model determined the identified risk equation as follows: Risk score = (0.2659 × PICSAR expression) + (0.4374 × FOXD2-AS1 expression) + (-0.3467 × AP001065.1 expression). The CupRLSig high-risk group was associated with poor overall survival (hazard ratio = 1.162, 95%CI = 1.063-1.270; P < 0.001) after the patients were divided into two groups depending upon their median risk score. Model accuracy was further supported by receiver operating characteristic and principal component analysis as well as the validation set. The area under the curve of 0.741 was found to be a better predictor of HCC prognosis as compared to other clinicopathological variables. Mutation analysis revealed that high-risk combinations with high TMB carried worse prognoses (median survival of 30 mo vs 102 mo of low-risk combinations with low TMB group). The low-risk group had more activated natural killer cells (NK cells, P = 0.032 by Wilcoxon rank sum test) and fewer regulatory T cells (Tregs, P = 0.021) infiltration than the high-risk group. This finding could explain why the low-risk group has a better prognosis. Interestingly, when checkpoint gene expression (CD276, CTLA-4, and PDCD-1) and tumor immune dysfunction and rejection (TIDE) scores are considered, high-risk patients may respond better to immunotherapy. Finally, most drugs commonly used in preclinical and clinical systemic therapy for HCC, such as 5-fluorouracil, gemcitabine, paclitaxel, imatinib, sunitinib, rapamycin, and XL-184 (cabozantinib), were found to be more efficacious in the low-risk group; erlotinib, an exception, was more efficacious in the high-risk group.
CONCLUSION The lncRNA signature, CupRLSig, constructed in this study is valuable in prognostic estimation of HCC. Importantly, CupRLSig also predicts the level of immune infiltration and potential efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- En-Min Huang
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Ning Ma
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Tao Ma
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Jun-Yi Zhou
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Wei-Sheng Yang
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Chuang-Xiong Liu
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Ze-Hui Hou
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Shuang Chen
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Zhen Zong
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Nanchang University, Nanchang 330006, Jiangxi Province, China
| | - Bing Zeng
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Ying-Ru Li
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| | - Tai-Cheng Zhou
- Department of Gastroenterological Surgery and Hernia Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, Guangdong Province, China
| |
Collapse
|
158
|
Karri S, Aviel-Ronen S, Firer MA. Fractal and textural imaging identify new subgroups of patients with colorectal cancer based on biophysical properties of the cancer cells. Pathol Res Pract 2022; 238:154040. [PMID: 36057191 DOI: 10.1016/j.prp.2022.154040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 10/16/2022]
Abstract
Colorectal cancer (CRC) can been sub-divided, based on the generation of tertiary lymphoid structures (TLS), into CRC with a Crohn's like lymphoid reaction (CLR) representing de novo formation of TLSs or CRC lacking TLSs that show Diffuse Inflammatory infiltration (DII). The association between TLS, early treatment initiation and longer survival highlights the need for deeper patient stratification that could lead to more targeted therapies. We hypothesized that such stratification might be achieved by using digital image analyses. Here we retrospectively analyzed 35 CRC patient samples classified as CLR or DII by digital analysis, focusing on the parameters Fractal dimension, Lacunarity and the textural features Angular second momentum, Correlation, Inverse difference momentum and Entropy. Significant differences in the grades of these parameters between the two patient groups provided preliminary data that additional biophysical information can divide CRC into at least 3 subgroups which encompass CLR and DII. Additional studies are needed to test if this sub-classification aids in the selection of targeted therapy for patients with CRC.
Collapse
Affiliation(s)
- Sirish Karri
- Department of Chemical Engineering, Ariel University, Ariel 40700, Israel
| | | | - Michael A Firer
- Department of Chemical Engineering, Ariel University, Ariel 40700, Israel; Adelson School of Medicine, Ariel University, Ariel 40700, Israel; Ariel Center for Applied Cancer Research, Ariel University, Ariel 40700, Israel.
| |
Collapse
|
159
|
Combination of Tumor Mutational Burden and DNA Damage Repair Gene Mutations with Stromal/Immune Scores Improved Prognosis Stratification in Patients with Lung Adenocarcinoma. JOURNAL OF ONCOLOGY 2022; 2022:6407344. [PMID: 36262349 PMCID: PMC9576425 DOI: 10.1155/2022/6407344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 07/24/2022] [Indexed: 12/24/2022]
Abstract
Background Both the tumor environment and the genomic landscape of lung cancer may shape patient responses to treatments, including immunotherapy, but their joint impacts on lung adenocarcinoma (LUAD) prognosis are underexplored. Methods RNA sequencing data and whole-exome sequencing results were downloaded from the TCGA database, and only LUAD-related data were included in this study. Based on gene expression data, the ESTIMATE algorithm was used to estimate stromal and immune scores, and CIBERSORT analysis was used for quantification of the relative abundances of immune cells. Somatic mutations were used for calculating tumor mutation burden (TMB). Specific mutations in genes involved in DNA damage repair (DDR) pathways were identified. The individual and joint associations of stromal and immune score, TMB, and DDR gene mutations with 5-year survival were analyzed by the Kaplan–Meier method and multivariate Cox model. Results LUAD patients with a high (>highest 25%) stromal or immune score had prolonged survival as compared to those with a low (<lowest 25%) score (log-rank P=0.05 and 0.035, respectively). Patients with both high stromal and immune scores had the most favorable survival. Although the survival differences between patients with high (>highest 25%) and low (<lowest 25%) TMB, or between patients with mutant- and wild-type DDR genes were not statistically significant, a survival benefit from high TMB or DDR gene mutations was observed in patients with high stromal or immune scores. Conclusion A comprehensive evaluation of transcriptomic signatures and genomic biomarkers may provide a novel avenue for improving prognosis stratification in LUAD.
Collapse
|
160
|
Wang J, Gu X, Cao L, Ouyang Y, Qi X, Wang Z, Wang J. A novel prognostic biomarker CD3G that correlates with the tumor microenvironment in cervical cancer. Front Oncol 2022; 12:979226. [PMID: 36176400 PMCID: PMC9513466 DOI: 10.3389/fonc.2022.979226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
Cervical cancer (CESC) is the fourth most common and death-causing gynecological cancer, mostly induced by infection of human papillomavirus (HPV). Multiple components of the tumor microenvironment (TME), such as tumor infiltrating immune cells, could be targets of immunotherapy for HPV-related CESC. However, little is known about the TME of CESC until now. Here, we aimed to uncover the pathogenesis as well as to identify novel biomarkers to predict prognosis and immunotherapy efficacy for CESC. Combining the transcriptomic data and clinical characteristics, we identified differentially expressed genes in CESC samples from TCGA database by comparing the two groups with different ImmuneScore and StromalScore. Next, we detected ten key genes based on the PPI network and survival analyses with the univariate Cox regression model. Thereafter, we focused on CD3G, the only gene exhibiting increased RNA and protein expression in tumors by multiple analyses. Higher CD3G expression was associated with better survival; and it was also significantly associated with immune-related pathways through GSEA analysis. Furthermore, we found that CD3G expression was correlated with 16 types of TICs. Single cell RNA-sequencing data of CD3G in lymphocytes subgroup indicated its possible role in HPV defense. Hence, CD3G might be a novel biomarker in prognosis and immunotherapy for CESC patients.
Collapse
Affiliation(s)
- Jingshuai Wang
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xuemin Gu
- Department of Obstetrics and Gynecology, Tongji Hospital of Tongji University, Shanghai, China
| | - Leilei Cao
- Department of Obstetrics and Gynecology, Shanghai Eighth People’s Hospital, Shanghai, China
| | - Yiqin Ouyang
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiao Qi
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhijie Wang
- Department of Obstetrics and Gynecology, Shanghai Eighth People’s Hospital, Shanghai, China
- *Correspondence: Jianjun Wang, ; Zhijie Wang,
| | - Jianjun Wang
- Department of Obstetrics and Gynecology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
- *Correspondence: Jianjun Wang, ; Zhijie Wang,
| |
Collapse
|
161
|
Mlecnik B, Torigoe T, Bindea G, Popivanova B, Xu M, Fujita T, Hazama S, Suzuki N, Nagano H, Okuno K, Hirohashi Y, Furuhata T, Takemasa I, Patel P, Vora H, Shah B, Patel JB, Rajvik KN, Pandya SJ, Shukla SN, Wang Y, Zhang G, Yoshino T, Taniguchi H, Bifulco C, Lugli A, Lee JKJ, Zlobec I, Rau TT, Berger MD, Nagtegaal ID, Vink-Börger E, Hartmann A, Geppert CI, Kolwelter J, Merkel S, Grützmann R, Van den Eynde M, Jouret-Mourin A, Kartheuser A, Léonard D, Remue C, Wang J, Bavi P, Roehrl MHA, Ohashi PS, Nguyen LT, Han S, MacGregor HL, Hafezi-Bakhtiari S, Wouters BG, Masucci GV, Andersson E, Zavadova E, Vocka M, Spacek J, Petruzelka L, Konopasek B, Dundr P, Skalova H, Nemejcova K, Botti G, Tatangelo F, Delrio P, Ciliberto G, Maio M, Laghi L, Grizzi F, Marliot F, Fredriksen T, Buttard B, Lafontaine L, Maby P, Majdi A, Hijazi A, El Sissy C, Kirilovsky A, Berger A, Lagorce C, Paustian C, Ballesteros-Merino C, Dijkstra J, Van de Water C, van Lent-van Vliet S, Knijn N, Mușină AM, Scripcariu DV, Marincola FM, Ascierto PA, Fox BA, Pagès F, Kawakami Y, Galon J. Clinical Performance of the Consensus Immunoscore in Colon Cancer in the Asian Population from the Multicenter International SITC Study. Cancers (Basel) 2022; 14:4346. [PMID: 36139506 PMCID: PMC9497086 DOI: 10.3390/cancers14184346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/30/2022] [Accepted: 09/01/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND: In this study, we evaluated the prognostic value of Immunoscore in patients with stage I−III colon cancer (CC) in the Asian population. These patients were originally included in an international study led by the Society for Immunotherapy of Cancer (SITC) on 2681 patients with AJCC/UICC-TNM stages I−III CC. METHODS: CD3+ and cytotoxic CD8+ T-lymphocyte densities were quantified in the tumor and invasive margin by digital pathology. The association of Immunoscore with prognosis was evaluated for time to recurrence (TTR), disease-free survival (DFS), and overall survival (OS). RESULTS: Immunoscore stratified Asian patients (n = 423) into different risk categories and was not impacted by age. Recurrence-free rates at 3 years were 78.5%, 85.2%, and 98.3% for a Low, Intermediate, and High Immunoscore, respectively (HR[Low-vs-High] = 7.26 (95% CI 1.75−30.19); p = 0.0064). A High Immunoscore showed a significant association with prolonged TTR, OS, and DFS (p < 0.05). In Cox multivariable analysis stratified by center, Immunoscore association with TTR was independent (HR[Low-vs-Int+High] = 2.22 (95% CI 1.10−4.55) p = 0.0269) of the patient’s gender, T-stage, N-stage, sidedness, and MSI status. A significant association of a High Immunoscore with prolonged TTR was also found among MSS (HR[Low-vs-Int+High] = 4.58 (95% CI 2.27−9.23); p ≤ 0.0001), stage II (HR[Low-vs-Int+High] = 2.72 (95% CI 1.35−5.51); p = 0.0052), low-risk stage-II (HR[Low-vs-Int+High] = 2.62 (95% CI 1.21−5.68); p = 0.0146), and high-risk stage II patients (HR[Low-vs-Int+High] = 3.11 (95% CI 1.39−6.91); p = 0.0055). CONCLUSION: A High Immunoscore is significantly associated with the prolonged survival of CC patients within the Asian population.
Collapse
Affiliation(s)
- Bernhard Mlecnik
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Inovarion, 75005 Paris, France
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Gabriela Bindea
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Boryana Popivanova
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Mingli Xu
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Tomonobu Fujita
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Shoichi Hazama
- Department of Translational Research and Developmental Therapeutics against Cancer, Yamaguchi University School of Medicine, Yamaguchi 755-8505, Japan
| | - Nobuaki Suzuki
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan
| | - Hiroaki Nagano
- Department of Gastroenterological, Breast and Endocrine Surgery, Yamaguchi University Graduate School of Medicine, Yamaguchi 753-8511, Japan
| | - Kiyotaka Okuno
- Department of Surgery, Kindai University, School of Medicine, Osakasayama 589-0014, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Tomohisa Furuhata
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Ichiro Takemasa
- Department of Surgery, Surgical Oncology, and Science, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Prabhudas Patel
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Hemangini Vora
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Birva Shah
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | | | - Kruti N. Rajvik
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Shashank J. Pandya
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Shilin N. Shukla
- The Gujarat Cancer & Research Institute, Asarwa, Ahmedabad 380016, India
| | - Yili Wang
- Institute for Cancer Research, School of Basic Medical Science, Xi’an 710061, China
- Health Science Center of Xi’an Jiaotong University, Xi’an 710061, China
| | - Guanjun Zhang
- Institute for Cancer Research, School of Basic Medical Science, Xi’an 710061, China
- Health Science Center of Xi’an Jiaotong University, Xi’an 710061, China
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwanoha, Kashiwa-shi 277-8577, Japan
| | - Hiroya Taniguchi
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwanoha, Kashiwa-shi 277-8577, Japan
| | - Carlo Bifulco
- Department of Pathology, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Alessandro Lugli
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Jiun-Kae Jack Lee
- Department of Biostatistics, M.D. Anderson Cancer Center, University of Texas, Houston, TX 77030, USA
| | - Inti Zlobec
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Tilman T. Rau
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland
| | - Martin D. Berger
- Department of Medical Oncology, University Hospital of Bern, 3010 Bern, Switzerland
| | - Iris D. Nagtegaal
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Elisa Vink-Börger
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Arndt Hartmann
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Carol I. Geppert
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Julie Kolwelter
- Department of Pathology, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Susanne Merkel
- Department of Surgery, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Grützmann
- Department of Surgery, University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marc Van den Eynde
- Institut Roi Albert II, Department of Medical Oncology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
- Institut de Recherche Clinique et Experimentale (Pole MIRO), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Anne Jouret-Mourin
- Department of Pathology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
- Institut de Recherche Clinique et Experimentale (Pole GAEN), Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Alex Kartheuser
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Daniel Léonard
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Christophe Remue
- Institut Roi Albert II, Department of Digestive Surgery, Cliniques Universitaires St-Luc Université Catholique de Louvain, 1200 Brussels, Belgium
| | - Julia Wang
- Curandis, New York, NY 10583, USA
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Prashant Bavi
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael H. A. Roehrl
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Linh T. Nguyen
- Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | - SeongJun Han
- Princess Margaret Cancer Centre, Toronto, ON M5G 2C1, Canada
| | | | - Sara Hafezi-Bakhtiari
- Department of Pathology, Laboratory Medicine Program, University Health Network, 11-E444, Toronto, ON M5G 2C4, Canada
| | | | - Giuseppe V. Masucci
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, 17177 Stockholm, Sweden
| | - Emilia Andersson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University, 17177 Stockholm, Sweden
| | - Eva Zavadova
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Michal Vocka
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Jan Spacek
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Lubos Petruzelka
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Bohuslav Konopasek
- Department of Oncology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Pavel Dundr
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Helena Skalova
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Kristyna Nemejcova
- Institute of Pathology, First Faculty of Medicine, General University Hospital in Prague, Charles University, 12808 Prague, Czech Republic
| | - Gerardo Botti
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Fabiana Tatangelo
- Department of Pathology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | - Paolo Delrio
- Colorectal Surgery Department, Instituto Nazionale Tumori IRCCS Fondazione G. Pascale, 80131 Naples, Italy
| | | | - Michele Maio
- Center for Immuno-Oncology, University Hospital, 53100 Siena, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, 20090 Milan, Italy
- Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy
| | - Fabio Grizzi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, 20090 Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Florence Marliot
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Tessa Fredriksen
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Bénédicte Buttard
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Lucie Lafontaine
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Pauline Maby
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Amine Majdi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Assia Hijazi
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| | - Carine El Sissy
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Amos Kirilovsky
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Anne Berger
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Digestive Surgery Department, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Christine Lagorce
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Department of Pathology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Christopher Paustian
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carmen Ballesteros-Merino
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Jeroen Dijkstra
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | | | | | - Nikki Knijn
- Pathology Department, Radboud University, 6500 HC Nijmegen, The Netherlands
| | - Ana-Maria Mușină
- Department of Surgical Oncology, Regional Institute of Oncology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iaşi, Romania
| | - Dragos-Viorel Scripcariu
- Department of Surgical Oncology, Regional Institute of Oncology, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iaşi, Romania
| | | | - Paolo A. Ascierto
- Melanoma, Cancer Immunotherapy and Innovative Therapies Unit, Istituto Nazionale Tumori IRCCS Fondazione “G. Pascale”, 80131 Naples, Italy
| | - Bernard A. Fox
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR 97239, USA
- Laboratory of Molecular and Tumor Immunology, Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR 97213, USA
| | - Franck Pagès
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
- Immunomonitoring Platform, Laboratory of Immunology, AP-HP, Assistance Publique-Hopitaux de Paris, Georges Pompidou European Hospital, 75015 Paris, France
| | - Yutaka Kawakami
- Division of Cellular Signaling, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, 75006 Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006 Paris, France
- Equipe Labellisée Ligue Contre le Cancer, 75006 Paris, France
| |
Collapse
|
162
|
Wu Y, Biswas D, Swanton C. Impact of cancer evolution on immune surveillance and checkpoint inhibitor response. Semin Cancer Biol 2022; 84:89-102. [PMID: 33631295 PMCID: PMC9253787 DOI: 10.1016/j.semcancer.2021.02.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/21/2022]
Abstract
Intratumour heterogeneity (ITH) is pervasive across all cancers studied and may provide the evolving tumour multiple routes to escape immune surveillance. Immune checkpoint inhibitors (CPIs) are rapidly becoming standard of care for many cancers. Here, we discuss recent work investigating the influence of ITH on patient response to immune checkpoint inhibitor (CPI) therapy. At its simplest, ITH may confound the diagnostic accuracy of predictive biomarkers used to stratify patients for CPI therapy. Furthermore, ITH is fuelled by mechanisms of genetic instability that can both engage immune surveillance and drive immune evasion. A greater appreciation of the interplay between ITH and the immune system may hold the key to increasing the proportion of patients experiencing durable responses from CPI therapy.
Collapse
Affiliation(s)
- Yin Wu
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK; Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, SE1 9RT, UK
| | - Dhruva Biswas
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK; Bill Lyons Informatics Centre, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, London, WC1E 6DD, UK.
| |
Collapse
|
163
|
Zhang W, Zhang Z, Lou S, Li D, Ma Z, Xue L. Efficacy, safety and predictors of combined fruquintinib with programmed death-1 inhibitors for advanced microsatellite-stable colorectal cancer: A retrospective study. Front Oncol 2022; 12:929342. [PMID: 36119532 PMCID: PMC9472213 DOI: 10.3389/fonc.2022.929342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background Research findings have revealed that combining anti-angiogenesis inhibitors with programmed death-1(PD-1) inhibitors can reverse the immunosuppressive tumor microenvironment and enhance the antitumor immune response. To explore the therapeutic options for breaking immune tolerance in microsatellite stability (MSS) or mismatch repair-proficiency (pMMR) advanced colorectal cancer (CRC), we assessed the efficacy, safety and predictors of the fruquintinib and PD-1 inhibitors combination in patients with MSS/pMMR advanced CRC in a real-world environment. Methods We conducted a single-center retrospective study by collecting relevant data on patients with MSS/pMMR advanced CRC who received fruquintinib coupled with PD-1 inhibitors in the First Affiliated Hospital of Zhengzhou University between August 2019 and November 2021, focusing on progression-free survival. Results We enrolled 110 eligible patients in this study between August 2019 and November 2021. At the deadline (January 20, 2022), 13 patients had objective responses. The objective response rate was 11.8% (13/110, 95% confidence interval [CI]: 6.4-18.2), the disease control rate was 70.0% (82/110, 95% CI: 60.9-78.2), and the progression-free survival was 5.4 months (95% CI: 4.0-6.8). Liver metastases (hazard ratio [HR]: 0.594, 95% CI: 0.363-0.973, P<0.05), alkaline phosphatase elevation (ALP>160U/L) (HR: 0.478, 95%CI: 0.241-0.948, P<0.05), fibrinogen elevation (FIB>4g/L) (HR: 0.517, 95% CI: 0.313-0.855, P<0.05), and an increase in the ALP level from the baseline after treatment (HR: 1.673, 95% CI: 1.040-2.690, P<0.05) were negative predictors of the progression-free survival. A total of 101 of 110 patients experienced treatment-related adverse events, including 14 who experienced grade 3 or above treatment-related adverse events, and no treatment-related deaths occurred. Hypertension was the most frequently encountered grade 3 treatment-related adverse event. Conclusion Fruquintinib combined with PD-1 inhibitors has antitumor activity and manageable safety in treating patients with MSS/pMMR advanced CRC. Liver metastases, ALP level and FIB level might be a prediction of the patient response to this therapy.
Collapse
|
164
|
Defining the Immune Checkpoint Landscape in Human Colorectal Cancer Highlights the Relevance of the TIGIT/CD155 Axis for Optimizing Immunotherapy. Cancers (Basel) 2022; 14:cancers14174261. [PMID: 36077799 PMCID: PMC9454990 DOI: 10.3390/cancers14174261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022] Open
Abstract
While immune checkpoint (IC) therapies, particularly those targeting the PD-1/PD-L1 axis, have revolutionized the treatment of melanoma and several other cancers, their effect remains very limited in colorectal cancer (CRC). To define a comprehensive landscape of ICs in the human CRC tumor microenvironment (TME), we evaluated, using multiparametric flow cytometry, their ex vivo expression via tumor-infiltrating lymphocytes (TILs) (n = 40 CRCs) as well as that of their respective ligands on tumor and myeloid cells (n = 29). Supervised flow cytometry analyses showed that (i) most CD3+ TILs expressed PD-1 and TIGIT and, to a lesser extent, Tim-3, Lag3 and NKG2A, and (ii) EpCAM+ tumor cells and CD11b+ myeloid cells differed in their IC ligand expression profile, with a strikingly high expression of CD155 by tumor cells. An in situ analysis of IC and their ligands using immunohistochemistry on paraffin sections of CRC confirmed the overexpression of TIGIT and its ligand, CD155, in the TME. Most interestingly, an unsupervised clustering analysis of IC co-expression on CD4+ and CD8+ TILs identified two tumor subgroups, named IChigh and IClow. Altogether, our findings highlight the TIGIT/CD155 axis as a potential target that could be used in combination IC therapy in CRC.
Collapse
|
165
|
Guo C, Zeng F, Liu H, Wang J, Huang X, Luo J. Establish immune-related gene prognostic index for esophageal cancer. Front Genet 2022; 13:956915. [PMID: 36035171 PMCID: PMC9401516 DOI: 10.3389/fgene.2022.956915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Background: Esophageal cancer is a tumor type with high invasiveness and low prognosis. As immunotherapy has been shown to improve the prognosis of esophageal cancer patients, we were interested in the establishment of an immune-associated gene prognostic index to effectively predict the prognosis of patients. Methods: To establish the immune-related gene prognostic index of esophageal cancer (EC), we screened 363 upregulated and 83 downregulated immune-related genes that were differentially expressed in EC compared to normal tissues. By multivariate Cox regression and weighted gene coexpression network analysis (WGCNA), we built a prognostic model based on eight immune-related genes (IRGs). We confirmed the prognostic model in both TCGA and GEO cohorts and found that the low-risk group had better overall survival than the high-risk group. Results: In this study, we identified 363 upregulated IRGs and 83 downregulated IRGs. Next, we found a prognostic model that was constructed with eight IRGs (OSM, CEACAM8, HSPA6, HSP90AB1, PCSK2, PLXNA1, TRIB2, and HMGB3) by multivariate Cox regression analysis and WGCNA. According to the Kaplan–Meier survival analysis results, the model we constructed can predict the prognosis of patients with esophageal cancer. This result can be verified by the Gene Expression Omnibus (GEO). Patients were divided into two groups with different outcomes. IRGPI-low patients had better overall survival than IRGPI-high patients.Conclusion: Our findings indicated the potential value of the IRGPI risk model for predicting the prognosis of EC patients.
Collapse
Affiliation(s)
- Caiyu Guo
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
- Department of Radiotherapy, Graduate School of Dalian Medical University, Dalian, China
| | - Fanye Zeng
- Second Department of Medical Oncology, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hui Liu
- School of Computer Science and Technology, Nanjing Tech University, Nanjing, China
| | - Jianlin Wang
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Xue Huang
- Department of Radiotherapy, Changzhou Tumor Hospital, Changzhou, China
- *Correspondence: Xue Huang, ; Judong Luo,
| | - Judong Luo
- Department of Radiotherapy, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, China
- *Correspondence: Xue Huang, ; Judong Luo,
| |
Collapse
|
166
|
Sinicrope FA, Shi Q, Catteau A, Poage GM, Zemla TJ, Mlecnik B, Benson AB, Gill S, Goldberg RM, Kahlenberg MS, Nair SG, Shields AF, Smyrk TC, Galon J, Alberts SR. Immunoscore Is Prognostic in Low-Risk and High-Risk Stage III Colon Carcinomas Treated With Adjuvant Infusional Fluorouracil, Leucovorin, and Oxaliplatin in a Phase III Trial. JCO Precis Oncol 2022; 6:e2200010. [PMID: 35952316 PMCID: PMC9384943 DOI: 10.1200/po.22.00010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/31/2022] [Accepted: 06/24/2022] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The recommended duration of adjuvant fluoropyrimidine and oxaliplatin chemotherapy for patients with stage III colon cancer is based on tumor classification into clinically low-risk (T1-3 N1) and high-risk (T4 or N2) groups. We determined whether Immunoscore can enhance prognostication within these risk groups. MATERIALS AND METHODS Patients with stage III colon carcinomas (N = 600) were randomly selected from the infusional fluorouracil, leucovorin, and oxaliplatin arm of adjuvant trial NCCTG N0147 (Alliance for Clinical Trials in Oncology). Tumors were evaluated for Immunoscore that quantifies CD3+ and CD8+ T-cell densities in the tumor center and invasive margin by digital image analysis. Disease-free survival (DFS) by Immunoscore was analyzed using a multivariable Cox regression model in each risk group with adjustment for covariates including KRAS, BRAFV600E, and mismatch repair status. RESULTS Of 559 cancers with Immunoscore data, 299 (53.5%) were classified as clinically low-risk (T1-3 N1) and 260 (46.5%) as clinically high-risk (T4 and/or N2). Among patients with low-risk tumors, those with Immunoscore-Low versus Immunoscore-High tumors had significantly worse 5-year DFS rates (77.5% v 91.8%; hazard ratio, 1.70; 95% CI, 1.03 to 2.79; P = .037). Among patients with high-risk tumors, those with Immunoscore-Low versus Immunoscore-High tumors also had significantly worse DFS (55.3% v 70.3%; hazard ratio, 1.65; 95% CI, 1.11 to 2.47; P = .013). Tumors that were low-risk/Immunoscore-Low had similar outcomes as did tumors that were high-risk/Immunoscore-High (P = .174). Prognostication was significantly improved in multivariable models where Immunoscore was added to clinical risk parameters and limited biomarkers (likelihood ratio test P = .0003). CONCLUSION Immunoscore can refine patient prognosis beyond clinical risk group classification, suggesting its potential utility for adjuvant decision making.
Collapse
Affiliation(s)
- Frank A. Sinicrope
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Qian Shi
- Alliance Statistics and Data Center, Rochester, MN
| | | | | | | | - Bernhard Mlecnik
- INSERM, UMRS 1138, Laboratory of Integrative Cancer Immunology, Universite Paris Descartes, Paris, France
- Inovarion, Paris, France
| | | | - Sharlene Gill
- British Columbia Cancer Agency—Vancouver Cancer Center, Vancouver, BC, Canada
| | | | | | | | | | - Thomas C. Smyrk
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Jerome Galon
- INSERM, UMRS 1138, Laboratory of Integrative Cancer Immunology, Universite Paris Descartes, Paris, France
| | - Steven R. Alberts
- Division of Oncology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
167
|
Kang H, Seo MK, Park B, Yoon SO, Koh YW, Kim D, Kim S. Characterizing intrinsic molecular features of the immune subtypes of salivary mucoepidermoid carcinoma. Transl Oncol 2022; 24:101496. [PMID: 35917642 PMCID: PMC9352547 DOI: 10.1016/j.tranon.2022.101496] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/18/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION Characterizing the tumor microenvironment (TME) and immune landscape of cancer has been a promising step towards discovering new therapeutic biomarkers and guiding precision medicine; however, its application in mucoepidermoid carcinoma (MEC) has been sparse. Here, we conducted a comprehensive study to understand the properties of the TME and immune profiles of MEC. METHOD 20 patients with MEC were collected from Yonsei Head and Neck Cancer Centre, Yonsei University, South Korea. Total RNA sequencing was conducted to determine gene expression profiles. Bioinformatic and immunoinformatic analyses were applied to characterize the TME and identify immunophenotypic subgroups, and to investigate the molecular features that explain the distinct phenotypes. RESULTS The MEC samples were subdivided into two groups, immune hot and immune cold, based on the heterogenous immune cell-infiltration and activation level. The immune-hot subgroup exhibited a higher level of immune activity, including T cell infiltration, cytolytic score, IFN-γ, antigen-presenting machinery, and immune modulator genes. Further characterizing molecular features of two subgroups, downregulation of lipid metabolic regulators, including MLXIPL and FASN, and the migration of chemokines and leukocytes were observed, respectively. And, Group-specific expression of immune checkpoint molecules, such as TIGIT, PD-L2, and CTLA-4, was observed in the immune-hot group, which can be exploited as a potential immunotherapeutic biomarker. CONCLUSIONS Immunophenotypically heterogeneous MEC subgroups analysis has shown distinctive molecular characteristics and provided potential treatment options. These findings yield new insights into TME of MEC and may help next step to study this uncharted cancer.
Collapse
Affiliation(s)
- Hyundeok Kang
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Mi-Kyoung Seo
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - BeumJin Park
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sun Och Yoon
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yoon Woo Koh
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dahee Kim
- Department of Otorhinolaryngology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|
168
|
Yunlong W, Tongtong L, Hua Z. The efficiency of neoadjuvant chemotherapy in colon cancer with mismatch repair deficiency. Cancer Med 2022; 12:2440-2452. [PMID: 35904113 PMCID: PMC9939115 DOI: 10.1002/cam4.5076] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/06/2022] [Accepted: 07/13/2022] [Indexed: 11/09/2022] Open
Abstract
Colon cancers with mismatch repair deficiency (dMMR) have specific clinicopathologic characteristics compared with mismatch repair proficiency (pMMR); however, the effect of MMR status on the efficiency of neoadjuvant chemotherapy (NCT) remains unclear. In our study, 439 dMMR and 26 pMMR colon cancer patients with or without NCT from 2010 to 2017 were retrospectively collected. Clinicopathological features, treatment response, and survival were compared between different groups. In the dMMR group, patients with NCT were likely to have higher CEA (abnormal CEA: 51.6% vs. 17.4%, p < 0.001), more multiorgan resection (38.7% vs. 16.8%, p = 0.006), and larger postoperative tumor diameter (7.26 vs. 6.21, p = 0.033) than those without NCT, but nearly half of cT4b patients who had NCT (42.9%, 9/21) avoid multiorgan resection. pT4 stage (HR, 14.97; 95% CI, 1.88-118.92; p = 0.010), number of positive lymph nodes (HR, 1.17; 95% CI, 1.09-1.26; p < 0.001), and tumor deposit (HR, 6.73; 95% CI, 2.08-21.74; p = 0.001) were independent prognosis factors of disease-free survival (DFS). For the advanced tumor subset, there is no significant difference between patients with or without NCT for OS (p = 0.13) and DFS (p = 0.11), although the survival rate of NCT was higher than non-NCT patients. Moreover, tumor regression grade was similar between dMMR and pMMR patients who had NCT. This study showed that NCT was more likely to be employed in dMMR patients with advanced tumors and may reduce the rate of multiorgan resection for cT4b dMMR patients. More large-scaled researches are needed to further explore if MMR status could predict the efficacy of neoadjuvant chemotherapy in patients with colon cancer.
Collapse
Affiliation(s)
- Wu Yunlong
- Department of General SurgeryBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Liu Tongtong
- Department of RadiologyBeijing Chao‐Yang Hospital, Capital Medical UniversityBeijingChina
| | - Zeng Hua
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
169
|
NK Cells and Other Cytotoxic Innate Lymphocytes in Colorectal Cancer Progression and Metastasis. Int J Mol Sci 2022; 23:ijms23147859. [PMID: 35887206 PMCID: PMC9322916 DOI: 10.3390/ijms23147859] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/12/2022] [Accepted: 07/14/2022] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies and leading causes of cancer-related deaths worldwide. Despite its complex pathogenesis and progression, CRC represents a well-fitting example of how the immune contexture can dictate the disease outcome. The presence of cytotoxic lymphocytes, both CD8+ T cells and natural killer (NK) cells, represents a relevant prognostic factor in CRC and is associated with a better overall survival. Together with NK cells, other innate lymphocytes, namely, innate lymphoid cells (ILCs), have been found both in biopsies of CRC patients and in murine models of intestinal cancer, playing both pro- and anti-tumor activities. In particular, several type 1 innate lymphoid cells (ILC1) with cytotoxic functions have been recently described, and evidence in mice shows a role for both NK cells and ILC1 in controlling CRC metastasis. In this review, we provide an overview of the features of NK cells and the expanding spectrum of innate lymphocytes with cytotoxic functions. We also comment on both the described and the potential roles these innate lymphocytes can play during the progression of intestinal cancer leading to metastasis. Finally, we discuss recent advances in the molecular mechanisms underlying the functional regulation of cytotoxic innate lymphocytes in CRC.
Collapse
|
170
|
Targeting interleukin-17 enhances tumor response to immune checkpoint inhibitors in colorectal cancer. Biochim Biophys Acta Rev Cancer 2022; 1877:188758. [PMID: 35809762 DOI: 10.1016/j.bbcan.2022.188758] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022]
Abstract
Although immune checkpoint inhibitors (ICIs) have gained much attention in managing cancer, only a minority of patients, especially those with tumors that have been classified as immunologically "cold" such as microsatellite stable (MSS) colorectal cancers (CRC), experience clinical benefit from ICIs. Surprisingly, interleukin-17 (IL-17) and its primary source Th17 are enriched in CRC and inversely associated with patient outcome. Our previous study revealed that IL-17A could upregulate programmed death-ligand 1 (PD-L1) expression and impede the efficacy of immunotherapy. IL-17, therefore, can be a possible target to sensitize tumor cells to ICIs. The detailed clinical results from our trial, which is the first to show the benefits of the combination of anti-PD-1 with anti-IL-17 therapy for MSS CRC, have also been presented. In this review, we highlight the role of IL-17 in ICIs resistance and summarize the current clinical evidence for the use of combination therapy. Directions for future strategies to warm up immunologically "cold" MSS CRCs have also been proposed.
Collapse
|
171
|
Mazzoli G, Cohen R, Lonardi S, Corti F, Elez E, Fakih M, Jayachandran P, Colle R, Shah AT, Salati M, Fenocchio E, Salvatore L, Ambrosini M, Ros J, Intini R, Cremolini C, Overman MJ, André T, Pietrantonio F. Prognostic impact of performance status on the outcomes of immune checkpoint inhibition strategies in patients with dMMR/MSI-H metastatic colorectal cancer. Eur J Cancer 2022; 172:171-181. [PMID: 35777274 DOI: 10.1016/j.ejca.2022.05.044] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/20/2022] [Accepted: 05/28/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors yielded unprecedented outcomes in patients with mismatch repair deficient/ microsatellite instability-high (dMMR/MSI-H) metastatic colorectal cancer (mCRC), but clinical decision-making in this rapidly evolving treatment landscape is challenging. Since performance status (PS) represents a well-established prognostic factor in clinical practice, we investigated whether worse PS, overall or related to either patients' frailty or high tumour burden, could affect the outcomes in this whole patients' population and according to immune checkpoint inhibitor treatment type. METHODS We conducted a global study at Tertiary Cancer Centres and collected data of patients with dMMR/MSI-H mCRC treated with anti- programmed-death (ligand)-1 (PD(L)-1) monotherapy or anti-PD-1/anti- cytotoxic T-lymphocyte antigen 4 combination. RESULTS The cohort included 502 patients. At a median follow-up of 31.2 months, worse PFS and OS were reported in patients with patient-related PS ≥ 1 (adjusted-HRs: 1.73, 95%CI: 1.06-2.83, p = 0.004 and 2.06, 95%CI: 1.13-3.74, p = 0.001, respectively) and cancer-related PS ≥ 1 (adjusted-HRs: 1.61, 95%CI: 1.19-2.17, p = 0.004 and 1.87, 95%CI: 1.32-2.66, p = 0.001, respectively). Anti-PD-1/anti- cytotoxic T-lymphocyte antigen 4 combination did not provide significantly better survival compared to anti-PD(L)-1 monotherapy in PS 0 subgroup (PFS HR = 0.62, 95%CI: 0.37-1.02, p = 0.059; OS HR = 0.59, 95%CI: 0.32-1.11, p = 0.100) and in patient-related PS ≥ 1 (PFS HR 0.93, 95%CI: 0.31-2.83, p = 0.899; OS HR 1.22, 95%CI: 0.34-4.37, p = 0.760), but the difference was significant and clinically meaningful in the subgroup with cancer-related PS ≥ 1 (PFS HR = 0.32, 95%CI: 0.19-0.53, p < 0.001; OS HR = 0.26, 95%CI: 0.14-0.48, p < 0.001). CONCLUSIONS In patients with dMMR/MSI-H mCRC, an extensive evaluation of clinical variables including PS may be implemented in the therapy decision-making.
Collapse
Affiliation(s)
- Giacomo Mazzoli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Romain Cohen
- Sorbonne Université, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP and INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Sara Lonardi
- Medical Oncology 3 and Medical Oncology 1, Istituto Oncologico Veneto IOV-IRCSS, Padua, Italy
| | - Francesca Corti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Elena Elez
- Department of Medical Oncology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutic Research, City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Priya Jayachandran
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Raphael Colle
- Sorbonne Université, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP and INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | | | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, PhD Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| | - Lisa Salvatore
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Università Cattolica del Sacro Cuore, Rome
| | - Margherita Ambrosini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan
| | - Javier Ros
- Department of Medical Oncology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Rossana Intini
- Medical Oncology 1, Istituto Oncologico Veneto IOV-IRCCS, Padua
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Michael J Overman
- Department of Gastrointestinal Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thierry André
- Sorbonne Université, Department of Medical Oncology, Hôpital Saint-Antoine, AP-HP and INSERM, Unité Mixte de Recherche Scientifique 938, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan.
| |
Collapse
|
172
|
Monteleone G, Maresca C, Colella M, Pacifico T, Congiu D, Troncone E, Marafini I. Targeting IL-34/MCSF-1R Axis in Colon Cancer. Front Immunol 2022; 13:917955. [PMID: 35837402 PMCID: PMC9273844 DOI: 10.3389/fimmu.2022.917955] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022] Open
Abstract
Colorectal carcinoma (CRC) is one of the most common neoplasias in the Western world and it is still one of the most deadly cancers worldwide mainly due to the fact that metastatic CRC is not responsive to current pharmacologic treatment. Identification of pathways that sustain CRC cell behaviour could help develop effective therapeutic compounds. A large body of evidence indicates that colon carcinogenesis is a dynamic process in which multiple cell types present in the tumor microenvironment either stimulate or suppress CRC cell growth, survival, and diffusion mainly via the production of cytokines. Interleukin-34 (IL-34), a cytokine initially known for its ability to regulate monocyte/macrophage survival and function, is highly produced in human CRC by both cancer cells and non-tumoral cells. IL-34 function is mainly mediated by interaction with the macrophage colony-stimulating factor-1 receptor (MCSF-1R), which is also over-expressed by CRC cells as well as by tumour-associated macrophages (TAMs) and cancer-associated fibroblasts. IL-34-driven MCSF-1R activation triggers several pro-tumoral functions in the colon. In this article, we review the current understanding of the involvement of IL-34 and its receptor in CRC, with particular attention to the available evidence about the IL-34/MCSF-1R axis-mediated regulation of TAMs and the role of IL-34 and MCSF-1R in promoting cancer resistance to chemotherapy and immunotherapy
Collapse
|
173
|
Bell PD, Pai RK. Immune Response in Colorectal Carcinoma: A Review of Its Significance as a Predictive and Prognostic Biomarker. Histopathology 2022; 81:696-714. [PMID: 35758208 DOI: 10.1111/his.14713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Colorectal carcinoma is a leading cause of cancer-related death worldwide. There is significant prognostic heterogeneity in stage II and III tumours, necessitating the development of new biomarkers to better identify patients at risk of disease progression. Recently, the tumour immune environment, particularly the type and quantity of T lymphocytes, has been shown to be a useful biomarker in predicting prognosis for patients with colorectal carcinoma. In this review, the significance of the immune response in colorectal carcinoma, including its influence on prognosis and response to therapy, will be detailed.
Collapse
Affiliation(s)
- Phoenix D Bell
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| | - Reetesh K Pai
- Department of Pathology, University of Pittsburgh Medical Centre, Pittsburgh, PA, 15213, USA
| |
Collapse
|
174
|
Martín-Otal C, Navarro F, Casares N, Lasarte-Cía A, Sánchez-Moreno I, Hervás-Stubbs S, Lozano T, Lasarte JJ. Impact of tumor microenvironment on adoptive T cell transfer activity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 370:1-31. [PMID: 35798502 DOI: 10.1016/bs.ircmb.2022.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Recent advances in immunotherapy have revolutionized the treatment of cancer. The use of adoptive cell therapies (ACT) such as those based on tumor infiltrating lymphocytes (TILs) or genetically modified cells (transgenic TCR lymphocytes or CAR-T cells), has shown impressive results in the treatment of several types of cancers. However, cancer cells can exploit mechanisms to escape from immunosurveillance resulting in many patients not responding to these therapies or respond only transiently. The failure of immunotherapy to achieve long-term tumor control is multifactorial. On the one hand, only a limited percentage of the transferred lymphocytes is capable of circulating through the bloodstream, interacting and crossing the tumor endothelium to infiltrate the tumor. Metabolic competition, excessive glucose consumption, the high level of lactic acid secretion and the extracellular pH acidification, the shortage of essential amino acids, the hypoxic conditions or the accumulation of fatty acids in the tumor microenvironment (TME), greatly hinder the anti-tumor activity of the immune cells in ACT therapy strategies. Therefore, there is a new trend in immunotherapy research that seeks to unravel the fundamental biology that underpins the response to therapy and identifies new approaches to better amplify the efficacy of immunotherapies. In this review we address important aspects that may significantly affect the efficacy of ACT, indicating also the therapeutic alternatives that are currently being implemented to overcome these drawbacks.
Collapse
Affiliation(s)
- Celia Martín-Otal
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Flor Navarro
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Noelia Casares
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Aritz Lasarte-Cía
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Inés Sánchez-Moreno
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Sandra Hervás-Stubbs
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Teresa Lozano
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
| | - Juan José Lasarte
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain.
| |
Collapse
|
175
|
Halima A, Vuong W, Chan TA. Next-generation sequencing: unraveling genetic mechanisms that shape cancer immunotherapy efficacy. J Clin Invest 2022; 132:154945. [PMID: 35703181 PMCID: PMC9197511 DOI: 10.1172/jci154945] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Immunity is governed by fundamental genetic processes. These processes shape the nature of immune cells and set the rules that dictate the myriad complex cellular interactions that power immune systems. Everything from the generation of T cell receptors and antibodies, control of epitope presentation, and recognition of pathogens by the immunoediting of cancer cells is, in large part, made possible by core genetic mechanisms and the cellular machinery that they encode. In the last decade, next-generation sequencing has been used to dissect the complexities of cancer immunity with potent effect. Sequencing of exomes and genomes has begun to reveal how the immune system recognizes “foreign” entities and distinguishes self from non-self, especially in the setting of cancer. High-throughput analyses of transcriptomes have revealed deep insights into how the tumor microenvironment affects immunotherapy efficacy. In this Review, we discuss how high-throughput sequencing has added to our understanding of how immune systems interact with cancer cells and how cancer immunotherapies work.
Collapse
Affiliation(s)
- Ahmed Halima
- Department of Radiation Oncology, Taussig Cancer Institute, and
| | - Winston Vuong
- Department of Radiation Oncology, Taussig Cancer Institute, and
| | - Timothy A Chan
- Department of Radiation Oncology, Taussig Cancer Institute, and.,Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio, USA.,National Center for Regenerative Medicine, Cleveland, Ohio, USA
| |
Collapse
|
176
|
Adaptive immune resistance at the tumour site: mechanisms and therapeutic opportunities. Nat Rev Drug Discov 2022; 21:529-540. [PMID: 35701637 DOI: 10.1038/s41573-022-00493-5] [Citation(s) in RCA: 207] [Impact Index Per Article: 69.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2022] [Indexed: 12/11/2022]
Abstract
Tumours employ various tactics to adapt and eventually resist immune attack. These mechanisms are collectively called adaptive immune resistance (AIR). The first defined and therapeutically validated AIR mechanism is the selective induction of programmed cell death 1 ligand 1 (PDL1) by interferon-γ in the tumour. Blockade of PDL1 binding to its receptor PD1 by antibodies (anti-PD therapy) has resulted in remission of a fraction of patients with advanced-stage cancer, especially in solid tumours. However, many clinical trials combining anti-PD therapy with other antitumour drugs conducted without a strong mechanistic rationale have failed to identify a synergistic or additive effect. In this Perspective article, we discuss why defining AIR mechanisms at the tumour site should be a key focus to direct future drug development as well as practical approaches to improve current cancer therapy.
Collapse
|
177
|
Waidhauser J, Nerlinger P, Sommer F, Wolf S, Eser S, Löhr P, Rank A, Märkl B. Circulating Lymphocytes Reflect the Local Immune Response in Patients with Colorectal Carcinoma. Diagnostics (Basel) 2022; 12:diagnostics12061408. [PMID: 35741218 PMCID: PMC9221878 DOI: 10.3390/diagnostics12061408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 12/04/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) correlate with the number and size of the surrounding lymph nodes in patients with colorectal carcinoma (CRC) and reflect the quality of the antitumor immune response. In this prospective study, we analyzed whether this response correlated with the circulating lymphocytes in peripheral blood (PB). In 47 patients with newly diagnosed CRC, flow cytometry was performed to analyze the B cells, T cells, NK cells, and a variety of their subsets in PB. The results were correlated with TILs in the resected tumor and with the number and size of the surrounding lymph nodes in nodal negative (N- patients (LN5: number of lymph nodes measuring ≥5 mm) and the metastasis-to-lymph node size ratio (MSR) in nodal positive patients (N+). Differences between the number of TILs could be seen between N+ and N- patients, dependent on the LN5 and MSR categories, with higher values in N- cases and in patients with a higher LN5 category or a lower MSR. Additionally, higher values of various circulating lymphocyte subgroups were observed in these patients. For the total PB lymphocytes, CD8 cells, and some of their subgroups, a positive correlation with the TILs was found. This study shows that circulating lymphocytes—in particular, cytotoxic T cells—correlate with the local antitumor immune response displayed by TILs and lymph node activation. Our findings indicate that local and generalized antitumor immune responses are concordant with their different components.
Collapse
Affiliation(s)
- Johanna Waidhauser
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
- Correspondence:
| | - Pia Nerlinger
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Florian Sommer
- Department of General, Visceral and Transplant Surgery, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (S.W.)
| | - Sebastian Wolf
- Department of General, Visceral and Transplant Surgery, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (F.S.); (S.W.)
| | - Stefan Eser
- Department of Gastroenterology and Infectious Diseases, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| | - Phillip Löhr
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Andreas Rank
- Department of Hematology and Oncology, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany; (P.N.); (P.L.); (A.R.)
| | - Bruno Märkl
- General Pathology and Molecular Diagnostics, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany;
| |
Collapse
|
178
|
Abstract
Skin cancers are among the most physically accessible malignancies, so local delivery of a medication into the tumor, so-called intratumoral therapy, is an appealing route of drug administration. Intratumoral therapies have the potential to increase local drug concentration and/or attract immune cells to the local tumor microenvironment, possibly with fewer systemic side effects. A wide array of intratumoral agents have been studied to date in patients with advanced melanoma, including chemotherapeutic drugs, immune modulating agents, and cancer-directed vaccines. In this review, we will summarize the key pre-clinical and clinical data supporting the use of intratumoral therapy for advanced unresectable and metastatic melanoma. First, we will discuss the history of intratumoral immunotherapy for the treatment of melanoma and the various agents studied to date. Second, we will explore how intratumoral therapies can constitute an in situ vaccine, potentially leading to disease control both locally and systemically. Finally, we will highlight opportunities in the field and key future directions.
Collapse
Affiliation(s)
- Laura A Huppert
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Adil I Daud
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
179
|
Singla B, Aithabathula RV, Kiran S, Kapil S, Kumar S, Singh UP. Reactive Oxygen Species in Regulating Lymphangiogenesis and Lymphatic Function. Cells 2022; 11:1750. [PMID: 35681445 PMCID: PMC9179518 DOI: 10.3390/cells11111750] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
The lymphatic system is pivotal for immunosurveillance and the maintenance of tissue homeostasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing vessels, has both physiological and pathological roles. Recent advances in the molecular mechanisms regulating lymphangiogenesis have opened a new area of research on reparative lymphangiogenesis for the treatment of various pathological disorders comprising neurological disorders, cardiac repair, autoimmune disease, obesity, atherosclerosis, etc. Reactive oxygen species (ROS) produced by the various cell types serve as signaling molecules in several cellular mechanisms and regulate various aspects of growth-factor-mediated responses, including lymphangiogenesis. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Low ROS levels are essential for lymphangiogenesis. On the contrary, oxidative stress due to enhanced ROS generation and/or reduced levels of antioxidants suppresses lymphangiogenesis via promoting lymphatic endothelial cell apoptosis and death. In this review article, we provide an overview of types and sources of ROS, discuss the role of ROS in governing lymphangiogenesis and lymphatic function, and summarize the role of lymphatics in various diseases.
Collapse
Affiliation(s)
- Bhupesh Singla
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Sonia Kiran
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Shweta Kapil
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children′s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| |
Collapse
|
180
|
Su X, Jin H, Wang J, Lu H, Gu T, Gao Z, Li M. Construction and validation of an immunoediting-based optimized neoantigen load (ioTNL) model to predict the response and prognosis of immune checkpoint therapy in various cancers. Aging (Albany NY) 2022; 14:4586-4605. [PMID: 35613927 PMCID: PMC9186755 DOI: 10.18632/aging.204101] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/12/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Only a minority of patients clinically benefit from immune checkpoint therapy. Tumor clones with neoantigens have immunogenicity; therefore, they are eliminated by T-cell-mediated immune editing. Identifying neoantigen clones with the ability to induce immune elimination may better predict the clinical outcome of immunotherapy. METHODS We developed ioTNL model, which indicates the immunoediting-based optimized tumor neoantigen load, by identifying tumor clones that could induce immune elimination. Data of more than two hundred patients from our patient pool and previously reported studies who underwent anti-PD-(L)1 therapy were collected to validate the prediction performance of ioTNL model. Clonal architectures, immune editing scores and ioTNL scores were identified. The association between the response as well as prognosis and the ioTNL were evaluated. Panel sequencing of genes from 2,469 patients within 20 cancer types was performed to profile the landscape of immunoediting. RESULTS As expected, the ioTNL score could predict the response in patients who underwent immune checkpoint inhibitor (ICI) immunotherapy for various cancers, including non-small cell lung cancer (NSCLC; p = 0.0066), skin cutaneous melanoma (SKCM; p = 0.026) and nasopharyngeal carcinoma (NPC; p = 0.0025). Patients with a high ioTNL score demonstrated longer survival than those with a low score. We verified the ioTNL on our cohort through panel sequencing and found that the ioTNL was associated with the response (p = 0.025) and prognosis (p = 0.00082) in anti-PD-(L)1 monotherapy. In addition, we found that the immune editing score correlated with the tumor mutation burden (TMB) and the objective response rate of immunotherapy. CONCLUSIONS Identifying neoantigen clones with the ability to induce immune elimination would better predict the efficacy of immunotherapy. We have proved that the reliable method of ioTNL can be applied to whole-exome sequencing (WES) and panel data and would have a broad application in precision diagnosis in immunotherapy.
Collapse
Affiliation(s)
- Xiaofan Su
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Haoxuan Jin
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Jiaqian Wang
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Huiping Lu
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Tiantian Gu
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Zhibo Gao
- YuceNeo Technology Co., Ltd., Shenzhen 518000, China
- YuceBio Technology Co., Ltd., Shenzhen 518020, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shanxi 710061, China
| |
Collapse
|
181
|
Regulatory T cells induce a suppressive immune milieu and promote lymph node metastasis in intrahepatic cholangiocarcinoma. Br J Cancer 2022; 127:757-765. [PMID: 35597869 PMCID: PMC9381563 DOI: 10.1038/s41416-022-01838-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/04/2022] Open
Abstract
Background Emerging evidence indicates that immunogenicity plays an important role in intrahepatic cholangiocarcinoma (ICC). Herein, we systematically evaluated the clinical relevance of immunogenicity in ICC. Methods Highly immunogenic ICCs identified in the public dataset and the Cancer Immunome Atlas (TCIA) were assessed to determine the prognostic impact of immunogenicity in ICC and key components after curative resection. We also investigated the clinical relevance of the immune milieu in ICC. Results Using the Gene Expression Omnibus dataset 89749 and TCIA, we identified CD8+/forkhead box P3 (FoxP3)+ tumour-infiltrating lymphocytes (TILs), T-cell immunoglobulin and mucin domain 3 (TIM-3) and human leukocyte antigen-A (HLA-A) in highly immunogenic ICCs. Immunohistochemical analysis of the in-house cohort showed that intratumoral FoxP3+ TILs correlated with CD8+ TILs (P = 0.045, Fisher’s exact test) and that high FoxP3+/CD8+ ratio (FCR) was an important marker for poor survival (P < 0.001, log-rank test). Furthermore, the FCR was higher in tumour-free lymph nodes in ICCs with lymph node metastases than in those without lymph node metastases (P = 0.003, Mann–Whitney U test). Conclusions FCR should be considered an important biomarker that represents the immune environment of ICC based on its potentially important role in tumour progression, especially lymph node metastasis.
Collapse
|
182
|
Sankar K, Ye JC, Li Z, Zheng L, Song W, Hu-Lieskovan S. The role of biomarkers in personalized immunotherapy. Biomark Res 2022; 10:32. [PMID: 35585623 PMCID: PMC9118650 DOI: 10.1186/s40364-022-00378-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/20/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors have revolutionized cancer therapeutic paradigm and substantially improved the survival of patients with advanced malignancies. However, a significant limitation is the wide variability in clinical response. MAIN TEXT Several biomarkers have been evaluated in prior and ongoing clinical trials to investigate their prognostic and predictive role of patient response, nonetheless, most have not been comprehensively incorporated into clinical practice. We reviewed published data regarding biomarkers that have been approved by the United States Food and Drug Administration as well as experimental tissue and peripheral blood biomarkers currently under investigation. We further discuss the role of current biomarkers to predict response and response to immune checkpoint inhibitors and the promise of combination biomarker strategies. Finally, we discuss ideal biomarker characteristics, and novel platforms for clinical trial design including enrichment and stratification strategies, all of which are exciting and dynamic to advance the field of precision immuno-oncology. CONCLUSION Incorporation and standardization of strategies to guide selection of combination biomarker approaches will facilitate expansion of the clinical benefit of immune checkpoint inhibitor therapy to appropriate subsets of cancer patients.
Collapse
Affiliation(s)
- Kamya Sankar
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Jing Christine Ye
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Lei Zheng
- Johns Hopkins University, Baltimore, MD, USA
| | - Wenru Song
- Kira Pharmaceuticals, Cambridge, MA, USA
| | - Siwen Hu-Lieskovan
- Division of Medical Oncology, University of Utah, Salt Lake City, UT, USA.
- Huntsman Cancer Institute, Salt Lake City, UT, USA.
| |
Collapse
|
183
|
Qian L, Lai X, Gu B, Sun X. An Immune-Related Gene Signature for Predicting Neoadjuvant Chemoradiotherapy Efficacy in Rectal Carcinoma. Front Immunol 2022; 13:784479. [PMID: 35603163 PMCID: PMC9121132 DOI: 10.3389/fimmu.2022.784479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 04/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background Locally advanced rectal cancers (LARC) show a highly variable response to neoadjuvant chemoradiotherapy (nCRT), and the impact of the tumor immune response in this process is poorly understood. This study aimed to characterize the immune-related gene expression profiles (GEP), pathways, and cell types associated with response or resistance to neoadjuvant chemoradiotherapy. Methods The transcriptomic and clinical data of Rectal carcinoma from the Gene Expression Omnibus database and Immune-related genes (IRGs) from ImmPort were downloaded to identify the differentially expressed immune-related genes (DEIRGs) between responder and non-responder to neoadjuvant chemoradiotherapy. Gene set enrichment analyses were performed to uncover significantly enriched GO terms and KEGG pathways. Immune cell infiltration was estimated from RNA-sequencing data using ImmuCellAI. Afterward, we constructed an immune-related gene-based predictive model (IRGPM) by Support Vector Machine and validated it in an external cohort. Result A 15-gene signature (HLA-DPB1, HLA-DQA1, CXCL9, CXCL10, TAP2, INHBB, BMP2, CD74, IL33, CCL11, CXCL11, DEFB1, HLA-DPA1, CCN3, STAT1) was identified as DEIRGs and found to be significantly associated with nCRT outcomes. Gene set enrichment analyses indicated that the 15 genes play active roles in inflammation-related biological processes. In addition, ImmuCellAI revealed that CD4 naive T cells, Tex, Th1 were significantly up-regulated (p=0.035, p=0.02, p=0.0086, respectively), while Tfh were significantly down-regulated (p=0.015) in responder subgroup. Finally, a novel predictive model was developed by SVM based on DEIRGs with an AUC of 80% (internal validation) and 73.5% (external validation). Conclusion Our team conducted a genomic study of the relationship between gene expression profile and response to nCRT in LARC. Our data suggested that the DEIRGs signature could help predict the efficacy of nCRT. And a DEIRGs‐based SVM model was developed to monitor the outcomes of nCRT in LARC.
Collapse
|
184
|
Dong J, Qian Y, Zhang G, Lu L, Zhang S, Ji G, Zhao A, Xu H. Can Natural Products be Used to Overcome the Limitations of Colorectal Cancer Immunotherapy? Front Oncol 2022; 12:884423. [PMID: 35600371 PMCID: PMC9114697 DOI: 10.3389/fonc.2022.884423] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/08/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is a common cancer of the digestive system that endangers human health. Immunotherapy is widely used in the treatment of patients with cancer. Some patients with dMMR/MSI-H CRC benefit from treatments that use immune checkpoint inhibitors, but most CRC patients are not sensitive to immunotherapy. Furthermore, internal resistance and immune escape lead to a reduced immunotherapy response. Therefore, the development of an effective combination therapy to improve the response rate to immunotherapy is a goal of cancer research. Natural products are potential candidates for comprehensive cancer treatments due to their wide range of immunomodulatory effects through multifactorial underlying mechanisms. In this review, we summarize the challenges in the treatment of CRC and assess the immunomodulatory effects of natural products and their active components. Our work suggests that natural products represent potential options for combined CRC immunotherapy.
Collapse
Affiliation(s)
- Jiahuan Dong
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yufan Qian
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guangtao Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lu Lu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shengan Zhang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Aiguang Zhao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
185
|
Vega PN, Nilsson A, Kumar MP, Niitsu H, Simmons AJ, Ro J, Wang J, Chen Z, Joughin BA, Li W, McKinley ET, Liu Q, Roland JT, Washington MK, Coffey RJ, Lauffenburger DA, Lau KS. Cancer-Associated Fibroblasts and Squamous Epithelial Cells Constitute a Unique Microenvironment in a Mouse Model of Inflammation-Induced Colon Cancer. Front Oncol 2022; 12:878920. [PMID: 35600339 PMCID: PMC9114773 DOI: 10.3389/fonc.2022.878920] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
The tumor microenvironment plays a key role in the pathogenesis of colorectal tumors and contains various cell types including epithelial, immune, and mesenchymal cells. Characterization of the interactions between these cell types is necessary for revealing the complex nature of tumors. In this study, we used single-cell RNA-seq (scRNA-seq) to compare the tumor microenvironments between a mouse model of sporadic colorectal adenoma (Lrig1CreERT2/+;Apc2lox14/+) and a mouse model of inflammation-driven colorectal cancer induced by azoxymethane and dextran sodium sulfate (AOM/DSS). While both models develop tumors in the distal colon, we found that the two tumor types have distinct microenvironments. AOM/DSS tumors have an increased abundance of two populations of cancer-associated fibroblasts (CAFs) compared with APC tumors, and we revealed their divergent spatial association with tumor cells using multiplex immunofluorescence (MxIF) imaging. We also identified a unique squamous cell population in AOM/DSS tumors, whose origins were distinct from anal squamous epithelial cells. These cells were in higher proportions upon administration of a chemotherapy regimen of 5-Fluorouracil/Irinotecan. We used computational inference algorithms to predict cell-cell communication mediated by ligand-receptor interactions and downstream pathway activation, and identified potential mechanistic connections between CAFs and tumor cells, as well as CAFs and squamous epithelial cells. This study provides important preclinical insight into the microenvironment of two distinct models of colorectal tumors and reveals unique roles for CAFs and squamous epithelial cells in the AOM/DSS model of inflammation-driven cancer.
Collapse
Affiliation(s)
- Paige N. Vega
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Avlant Nilsson
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Manu P. Kumar
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Hiroaki Niitsu
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Alan J. Simmons
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James Ro
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Jiawei Wang
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Zhengyi Chen
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Brian A. Joughin
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Wei Li
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Eliot T. McKinley
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Qi Liu
- Department of Biostatistics and Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Joseph T. Roland
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| | - M. Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Robert J. Coffey
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Douglas A. Lauffenburger
- Department of Biological Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Ken S. Lau
- Department of Cell and Developmental Biology and Program in Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
186
|
Haag GM, Springfeld C, Grün B, Apostolidis L, Zschäbitz S, Dietrich M, Berger AK, Weber TF, Zoernig I, Schaaf M, Waberer L, Müller DW, Al-Batran SE, Halama N, Jaeger D. Pembrolizumab and maraviroc in refractory mismatch repair proficient/microsatellite-stable metastatic colorectal cancer – The PICCASSO phase I trial. Eur J Cancer 2022; 167:112-122. [DOI: 10.1016/j.ejca.2022.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/13/2022] [Indexed: 12/20/2022]
|
187
|
Zimmer CL, Filipovic I, Cornillet M, O'Rourke CJ, Berglin L, Jansson H, Sun D, Strauss O, Hertwig L, Johansson H, von Seth E, Sparrelid E, Dias J, Glaumann H, Melum E, Ellis EC, Sandberg JK, Andersen JB, Bergquist A, Björkström NK. Mucosal-associated invariant T-cell tumor infiltration predicts long-term survival in cholangiocarcinoma. Hepatology 2022; 75:1154-1168. [PMID: 34719787 DOI: 10.1002/hep.32222] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/17/2021] [Accepted: 10/28/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Cholangiocarcinoma (CCA) is a malignancy arising from biliary epithelial cells of intra- and extrahepatic bile ducts with dismal prognosis and few nonsurgical treatments available. Despite recent success in the immunotherapy-based treatment of many tumor types, this has not been successfully translated to CCA. Mucosal-associated invariant T (MAIT) cells are cytotoxic innate-like T cells highly enriched in the human liver, where they are located in close proximity to the biliary epithelium. Here, we aimed to comprehensively characterize MAIT cells in intrahepatic (iCCA) and perihilar CCA (pCCA). APPROACH AND RESULTS Liver tissue from patients with CCA was used to study immune cells, including MAIT cells, in tumor-affected and surrounding tissue by immunohistochemistry, RNA-sequencing, and multicolor flow cytometry. The iCCA and pCCA tumor microenvironment was characterized by the presence of both cytotoxic T cells and high numbers of regulatory T cells. In contrast, MAIT cells were heterogenously lost from tumors compared to the surrounding liver tissue. This loss possibly occurred in response to increased bacterial burden within tumors. The residual intratumoral MAIT cell population exhibited phenotypic and transcriptomic alterations, but a preserved receptor repertoire for interaction with tumor cells. Finally, the high presence of MAIT cells in livers of iCCA patients predicted long-term survival in two independent cohorts and was associated with a favorable antitumor immune signature. CONCLUSIONS MAIT cell tumor infiltration associates with favorable immunological fitness and predicts survival in CCA.
Collapse
Affiliation(s)
- Christine L Zimmer
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Iva Filipovic
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Martin Cornillet
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Colm J O'Rourke
- Biotech Research and Innovation Centre (BRIC)Department of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Lena Berglin
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Hannes Jansson
- Division of SurgeryDepartment of Clinical Science, Intervention and TechnologyKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Dan Sun
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Otto Strauss
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Laura Hertwig
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Helene Johansson
- Division of Transplantation SurgeryCLINTECKarolinka Institutet and Department of TransplantationKarolinska University HospitalStockholmSweden
| | - Erik von Seth
- Division of Upper GI DiseasesKarolinska University HospitalStockholmSweden
- Unit of Gastroenterology and RheumatologyDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Ernesto Sparrelid
- Division of SurgeryDepartment of Clinical Science, Intervention and TechnologyKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Joana Dias
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Hans Glaumann
- Department of MedicineClinical Pathology and CytologyKarolinska University HospitalStockholmSweden
| | - Espen Melum
- Norwegian PSC Research CenterDepartment of Transplantation MedicineDivision of SurgeryInflammatory Diseases and TransplantationOslo University Hospital RikshospitaletOsloNorway
- Research Institute of Internal MedicineDivision of SurgeryInflammatory Diseases and TransplantationOslo University HospitalOsloNorway
- Institute of Clinical MedicineFaculty of MedicineUniversity of OsloOsloNorway
- Section of GastroenterologyDepartment of Transplantation MedicineDivision of SurgeryInflammatory Diseases and TransplantationOslo University Hospital RikshospitaletOsloNorway
- Hybrid Technology Hub-Centre of ExcellenceInstitute of Basic Medical SciencesFaculty of MedicineUniversity of OsloOsloNorway
| | - Ewa C Ellis
- Division of Transplantation SurgeryCLINTECKarolinka Institutet and Department of TransplantationKarolinska University HospitalStockholmSweden
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC)Department of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Annika Bergquist
- Division of Upper GI DiseasesKarolinska University HospitalStockholmSweden
- Unit of Gastroenterology and RheumatologyDepartment of Medicine HuddingeKarolinska InstitutetKarolinska University HospitalStockholmSweden
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine HuddingeKarolinska Institutet, Karolinska University HospitalStockholmSweden
| |
Collapse
|
188
|
Lahoz S, Archilla I, Asensio E, Hernández‐Illán E, Ferrer Q, López‐Prades S, Nadeu F, Del Rey J, Sanz‐Pamplona R, Lozano JJ, Castells A, Cuatrecasas M, Camps J. Copy-number intratumor heterogeneity increases the risk of relapse in chemotherapy-naive stage II colon cancer. J Pathol 2022; 257:68-81. [PMID: 35066875 PMCID: PMC9790656 DOI: 10.1002/path.5870] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/17/2021] [Accepted: 01/13/2022] [Indexed: 12/30/2022]
Abstract
Optimal selection of high-risk patients with stage II colon cancer is crucial to ensure clinical benefit of adjuvant chemotherapy. Here, we investigated the prognostic value of genomic intratumor heterogeneity and aneuploidy for disease recurrence. We combined targeted sequencing, SNP arrays, fluorescence in situ hybridization, and immunohistochemistry on a retrospective cohort of 84 untreated stage II colon cancer patients. We assessed the clonality of copy-number alterations (CNAs) and mutations, CD8+ lymphocyte infiltration, and their association with time to recurrence. Prognostic factors were included in machine learning analysis to evaluate their ability to predict individual relapse risk. Tumors from recurrent patients displayed a greater proportion of CNAs compared with non-recurrent (mean 31.3% versus 23%, respectively; p = 0.014). Furthermore, patients with elevated tumor CNA load exhibited a higher risk of recurrence compared with those with low levels [p = 0.038; hazard ratio (HR) 2.46], which was confirmed in an independent cohort (p = 0.004; HR 3.82). Candidate chromosome-specific aberrations frequently observed in recurrent cases included gain of the chromosome arm 13q (p = 0.02; HR 2.67) and loss of heterozygosity at 17q22-q24.3 (p = 0.05; HR 2.69). CNA load positively correlated with intratumor heterogeneity (R = 0.52; p < 0.0001). Consistently, incremental subclonal CNAs were associated with an elevated risk of relapse (p = 0.028; HR 2.20), which we did not observe for subclonal single-nucleotide variants and small insertions and deletions. The clinico-genomic model rated an area under the curve of 0.83, achieving a 10% incremental gain compared with clinicopathological markers (p = 0.047). In conclusion, tumor aneuploidy and copy-number intratumor heterogeneity were predictive of poor outcome and improved discriminative performance in early-stage colon cancer. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Sara Lahoz
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Ivan Archilla
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Elena Asensio
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Eva Hernández‐Illán
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Queralt Ferrer
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Sandra López‐Prades
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Ferran Nadeu
- Molecular Pathology of Lymphoid NeoplasmsInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)BarcelonaSpain
| | - Javier Del Rey
- Department of Cell Biology, Physiology and Immunology, Faculty of MedicineUniversity Autonomous of BarcelonaBellaterraSpain
| | - Rebeca Sanz‐Pamplona
- Unit of Biomarkers and SusceptibilityOncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL) and CIBERESPl'Hospitalet de LlobregatSpain
| | - Juan José Lozano
- Bioinformatics PlatformCentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD)MadridSpain
| | - Antoni Castells
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Miriam Cuatrecasas
- Pathology Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain
| | - Jordi Camps
- Translational Colorectal Cancer Genomics, Gastrointestinal and Pancreatic Oncology TeamInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), University of BarcelonaBarcelonaSpain,Department of Cell Biology, Physiology and Immunology, Faculty of MedicineUniversity Autonomous of BarcelonaBellaterraSpain
| |
Collapse
|
189
|
Huyghe N, Benidovskaya E, Stevens P, Van den Eynde M. Biomarkers of Response and Resistance to Immunotherapy in Microsatellite Stable Colorectal Cancer: Toward a New Personalized Medicine. Cancers (Basel) 2022; 14:2241. [PMID: 35565369 PMCID: PMC9105843 DOI: 10.3390/cancers14092241] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022] Open
Abstract
Immune Checkpoint Inhibitors (ICIs) are well recognized as a major immune treatment modality for multiple types of solid cancers. However, for colorectal cancer (CRC), ICIs are only approved for the treatment of Mismatch-Repair-Deficient and Microsatellite Instability-High (dMMR/MSI-H) tumors. For the vast majority of CRC, that are not dMMR/MSI-H, ICIs alone provide limited to no clinical benefit. This discrepancy of response between CRC and other solid cancers suggests that CRC may be inherently resistant to ICIs alone. In translational research, efforts are underway to thoroughly characterize the immune microenvironment of CRC to better understand the mechanisms behind this resistance and to find new biomarkers of response. In the clinic, trials are being set up to study biomarkers along with treatments targeting newly discovered immune checkpoint molecules or treatments combining ICIs with other existing therapies to improve response in MSS CRC. In this review, we will focus on the characteristics of response and resistance to ICIs in CRC, and discuss promising biomarkers studied in recent clinical trials combining ICIs with other therapies.
Collapse
Affiliation(s)
- Nicolas Huyghe
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Elena Benidovskaya
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Philippe Stevens
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
| | - Marc Van den Eynde
- Institut de Recherche Clinique et Expérimentale (Pole MIRO), UCLouvain, 1200 Brussels, Belgium; (N.H.); (E.B.); (P.S.)
- Institut Roi Albert II, Department of Medical Oncology and Gastroenterology, Cliniques Universitaires St-Luc, 1200 Brussels, Belgium
| |
Collapse
|
190
|
Abdeljaoued S, Arfa S, Kroemer M, Ben Khelil M, Vienot A, Heyd B, Loyon R, Doussot A, Borg C. Tissue-resident memory T cells in gastrointestinal cancer immunology and immunotherapy: ready for prime time? J Immunother Cancer 2022; 10:jitc-2021-003472. [PMID: 35470231 PMCID: PMC9039405 DOI: 10.1136/jitc-2021-003472] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2022] [Indexed: 12/12/2022] Open
Abstract
Tissue-resident memory T (TRM) cells have emerged as immune sentinels that patrol the tissue microenvironment and orchestrate localized antitumor immunity in various solid cancers. Recent studies have revealed that TRM cells are key players in cancer immunosurveillance, and their involvement has been linked to favorable responses to immunotherapy as well as general better clinical outcome in cancer patients. In this review, we provide an overview of the major advances and recent findings regarding TRM cells phenotype, transcriptional and epigenetic regulation in cancer with a special focus on gastrointestinal tumors. Finally, we highlight the exciting clinical implication of TRM cells in these types of tumors.
Collapse
Affiliation(s)
- Syrine Abdeljaoued
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France .,Clinical Investigational Center, CIC-1431, Besançon, France
| | - Sara Arfa
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Marie Kroemer
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Clinical Investigational Center, CIC-1431, Besançon, France.,Department of Pharmacy, University Hospital of Besançon, Besançon, France
| | - Myriam Ben Khelil
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France
| | - Angélique Vienot
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| | - Bruno Heyd
- Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Romain Loyon
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France
| | - Alexandre Doussot
- Department of Digestive and Oncologic Surgery, Liver Transplantation Unit, University Hospital of Besançon, Besançon, France
| | - Christophe Borg
- RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Besançon, France.,Clinical Investigational Center, CIC-1431, Besançon, France.,Department of Medical Oncology, University Hospital of Besançon, Besançon, France
| |
Collapse
|
191
|
Shinohara S, Takahashi Y, Komuro H, Matsui T, Sugita Y, Demachi-Okamura A, Muraoka D, Takahara H, Nakada T, Sakakura N, Masago K, Miyai M, Nishida R, Shomura S, Shigematsu Y, Hatooka S, Sasano H, Watanabe F, Adachi K, Fujinaga K, Kaneda S, Takao M, Ohtsuka T, Yamaguchi R, Kuroda H, Matsushita H. New evaluation of the tumor immune microenvironment of non-small cell lung cancer and its association with prognosis. J Immunother Cancer 2022; 10:jitc-2021-003765. [PMID: 35396225 PMCID: PMC8996063 DOI: 10.1136/jitc-2021-003765] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND A better understanding of the tumor immune microenvironment (TIME) will facilitate the development of prognostic biomarkers and more effective therapeutic strategies in patients with lung cancer. However, little has been reported on the comprehensive evaluation of complex interactions among cancer cells, immune cells, and local immunosuppressive elements in the TIME. METHODS Whole-exome sequencing and RNA sequencing were carried out on 113 lung cancers. We performed single sample gene set enrichment analysis on TIME-related gene sets to develop a new scoring system (TIME score), consisting of T-score (tumor proliferation), I-score (antitumor immunity) and S-score (immunosuppression). Lung cancers were classified according to a combination of high or low T-score, I-score, and S-scores (eight groups; G1-8). Clinical and genomic features, and immune landscape were investigated among eight groups. The external data sets of 990 lung cancers from The Cancer Genome Atlas and 76 melanomas treated with immune checkpoint inhibitors (ICI) were utilized to evaluate TIME scoring and explore prognostic and predictive accuracy. RESULTS The representative histological type including adenocarcinoma and squamous cell carcinoma, and driver mutations such as epidermal growth factor receptor and TP53 mutations were different according to the T-score. The numbers of somatic mutations and predicted neoantigens were higher in Thi (G5-8) than Tlo (G1-4) tumors. Immune selection pressure against neoantigen expression occurred only in Thi and was dampened in Thi/Ilo (G5-6), possibly due to a reduced number of T cells with a high proportion of tumor specific but exhausted cells. Thi/Ilo/Shi (G5) displayed the lowest immune responses by additional immune suppressive mechanisms. The T-score, I-score and S-scores were independent prognostic factors, with survival curves well separated into eight groups with G5 displaying the worst overall survival, while the opposite group Tlo/Ihi/Slo (G4) had the best prognosis. Several oncogenic signaling pathways influenced on T-score and I-scores but not S-score, and PI3K pathway alteration correlated with poor prognosis in accordance with higher T-score and lower I-score. Moreover, the TIME score predicted the efficacy of ICI in patients with melanoma. CONCLUSION The TIME score capturing complex interactions among tumor proliferation, antitumor immunity and immunosuppression could be useful for prognostic predictions or selection of treatment strategies in patients with lung cancer.
Collapse
Affiliation(s)
- Shuichi Shinohara
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Takahashi
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hiroyasu Komuro
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takuya Matsui
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yusuke Sugita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hirotomo Takahara
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takeo Nakada
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Noriaki Sakakura
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Katsuhiro Masago
- Pathology and Molecular Diagnostics, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Manami Miyai
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Reina Nishida
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Shin Shomura
- Department of Thoracic Surgery, Mie Prefectural General Medical Center, Yokkaichi, Japan
| | - Yoshiki Shigematsu
- Department of Respiratory Surgery, Ichinomiya Nishi Hospital, Ichinomiya, Japan
| | - Shunzo Hatooka
- Department of Respiratory Surgery, Ichinomiya Nishi Hospital, Ichinomiya, Japan
| | - Hajime Sasano
- Department of Respiratory Medicine and Allergy, Tosei General Hospital, Seto, Japan
| | - Fumiaki Watanabe
- Department of Thoracic Surgery, Mie Chuo Medical Center, Tsu, Japan
| | | | - Kazuya Fujinaga
- Department of Thoracic Surgery, Anjo Kosei Hospital, Anjo, Japan
| | - Shinji Kaneda
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Motoshi Takao
- Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Takashi Ohtsuka
- Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan.,Division of Cancer Informatics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kuroda
- Department of Thoracic Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan .,Division of Cancer Immunogenomics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
192
|
Lin Y, Kong DX, Zhang YN. Does the Microbiota Composition Influence the Efficacy of Colorectal Cancer Immunotherapy? Front Oncol 2022; 12:852194. [PMID: 35463305 PMCID: PMC9023803 DOI: 10.3389/fonc.2022.852194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is the second most common malignancy globally, and many people with CRC suffer the fate of death. Due to the importance of CRC and its negative impact on communities, treatment strategies to control it or increase patient survival are being studied. Traditional therapies, including surgery and chemotherapy, have treated CRC patients. However, with the advancement of science, we are witnessing the emergence of novel therapeutic approaches such as immunotherapy for CRC treatment, which have had relatively satisfactory clinical outcomes. Evidence shows that gastrointestinal (GI) microbiota, including various bacterial species, viruses, and fungi, can affect various biological events, regulate the immune system, and even treat diseases like human malignancies. CRC has recently shown that the gut microorganism pattern can alter both antitumor and pro-tumor responses, as well as cancer immunotherapy. Of course, this is also true of traditional therapies because it has been revealed that gut microbiota can also reduce the side effects of chemotherapy. Therefore, this review summarized the effects of gut microbiota on CRC immunotherapy.
Collapse
Affiliation(s)
- Yan Lin
- Health Management Center, Department of General Practice, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
- *Correspondence: Yan Lin, ; You-Ni Zhang,
| | - De-Xia Kong
- Health Management Center, Department of General Practice, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| | - You-Ni Zhang
- Department of Laboratory Medicine, Tiantai People’s Hospital, Taizhou, China
- *Correspondence: Yan Lin, ; You-Ni Zhang,
| |
Collapse
|
193
|
Masuda K, Kornberg A, Miller J, Lin S, Suek N, Botella T, Secener KA, Bacarella AM, Cheng L, Ingham M, Rosario V, Al-Mazrou AM, Lee-Kong SA, Kiran RP, Stoeckius M, Smibert P, Del Portillo A, Oberstein PE, Sims PA, Yan KS, Han A. Multiplexed single-cell analysis reveals prognostic and nonprognostic T cell types in human colorectal cancer. JCI Insight 2022; 7:e154646. [PMID: 35192548 PMCID: PMC9057629 DOI: 10.1172/jci.insight.154646] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/16/2022] [Indexed: 01/21/2023] Open
Abstract
Clinical outcomes in colorectal cancer (CRC) correlate with T cell infiltrates, but the specific contributions of heterogenous T cell types remain unclear. To investigate the diverse function of T cells in CRC, we profiled 37,931 T cells from tumors and adjacent normal colon of 16 patients with CRC with respect to transcriptome, TCR sequence, and cell surface markers. Our analysis identified phenotypically and functionally distinguishable effector T cell types. We employed single-cell gene signatures from these T cell subsets to query the TCGA database to assess their prognostic significance. We found 2 distinct cytotoxic T cell types. GZMK+KLRG1+ cytotoxic T cells were enriched in CRC patients with good outcomes. GNLY+CD103+ cytotoxic T cells with a dysfunctional phenotype were not associated with good outcomes, despite coexpression of CD39 and CD103, markers that denote tumor reactivity. We found 2 distinct Treg subtypes associated with opposite outcomes. While total Tregs were associated with good outcomes, CD38+ Tregs were associated with bad outcomes independently of stage and possessed a highly suppressive phenotype, suggesting that they inhibit antitumor immunity in CRC. These findings highlight the potential utility of these subpopulations in predicting outcomes and support the potential for novel therapies directed at CD38+ Tregs or CD8+CD103+ T cells.
Collapse
Affiliation(s)
| | - Adam Kornberg
- Columbia Center for Translational Immunology
- Department of Microbiology & Immunology
| | - Jonathan Miller
- Department of Pediatrics
- Columbia Center for Human Development
| | - Sijie Lin
- Columbia Center for Translational Immunology
| | - Nathan Suek
- Columbia Center for Translational Immunology
| | | | | | | | | | - Matthew Ingham
- Department of Medicine, Division of Hematology & Oncology
- Herbert Irving Comprehensive Cancer Center, and
| | - Vilma Rosario
- Herbert Irving Comprehensive Cancer Center, and
- Department of Surgery, Division of Colorectal Surgery, Columbia University, New York, New York, USA
| | - Ahmed M. Al-Mazrou
- Herbert Irving Comprehensive Cancer Center, and
- Department of Surgery, Division of Colorectal Surgery, Columbia University, New York, New York, USA
| | - Steven A. Lee-Kong
- Herbert Irving Comprehensive Cancer Center, and
- Department of Surgery, Division of Colorectal Surgery, Columbia University, New York, New York, USA
| | - Ravi P. Kiran
- Herbert Irving Comprehensive Cancer Center, and
- Department of Surgery, Division of Colorectal Surgery, Columbia University, New York, New York, USA
| | | | | | | | - Paul E. Oberstein
- Department of Medicine, Division of Hematology & Oncology
- Herbert Irving Comprehensive Cancer Center, and
| | - Peter A. Sims
- Departments of Systems Biology and Biochemistry & Molecular Biophysics
| | - Kelley S. Yan
- Columbia Center for Human Development
- Department of Medicine, Division of Digestive & Liver Diseases, and
- Department of Genetics & Development, Columbia University, New York, New York, USA
| | - Arnold Han
- Columbia Center for Translational Immunology
- Department of Microbiology & Immunology
- Herbert Irving Comprehensive Cancer Center, and
- Department of Medicine, Division of Digestive & Liver Diseases, and
| |
Collapse
|
194
|
Qi J, Sun H, Zhang Y, Wang Z, Xun Z, Li Z, Ding X, Bao R, Hong L, Jia W, Fang F, Liu H, Chen L, Zhong J, Zou D, Liu L, Han L, Ginhoux F, Liu Y, Ye Y, Su B. Single-cell and spatial analysis reveal interaction of FAP + fibroblasts and SPP1 + macrophages in colorectal cancer. Nat Commun 2022; 13:1742. [PMID: 35365629 PMCID: PMC8976074 DOI: 10.1038/s41467-022-29366-6] [Citation(s) in RCA: 444] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 03/11/2022] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the most common malignancies with limited treatments other than surgery. The tumor microenvironment (TME) profiling enables the discovery of potential therapeutic targets. Here, we profile 54,103 cells from tumor and adjacent tissues to characterize cellular composition and elucidate the potential origin and regulation of tumor-enriched cell types in CRC. We demonstrate that the tumor-specific FAP+ fibroblasts and SPP1+ macrophages were positively correlated in 14 independent CRC cohorts containing 2550 samples and validate their close localization by immuno-fluorescent staining and spatial transcriptomics. This interaction might be regulated by chemerin, TGF-β, and interleukin-1, which would stimulate the formation of immune-excluded desmoplasic structure and limit the T cell infiltration. Furthermore, we find patients with high FAP or SPP1 expression achieved less therapeutic benefit from an anti-PD-L1 therapy cohort. Our results provide a potential therapeutic strategy by disrupting FAP+ fibroblasts and SPP1+ macrophages interaction to improve immunotherapy. Tumour microenvironment profiling during colorectal cancer progression may enable the discovery of therapeutic targets. Here, single cell and spatial RNA sequencing of tumour and adjacent normal tissues reveals an interaction between FAP+ fibroblasts and SPP1+ macrophages that could be disrupted as an immunotherapy strategy.
Collapse
Affiliation(s)
- Jingjing Qi
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Biliary and Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongxiang Sun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zhang
- Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengting Wang
- Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenzhen Xun
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziyi Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyu Ding
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rujuan Bao
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liwen Hong
- Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenqing Jia
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Fang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongzhi Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhong
- Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Duowu Zou
- Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Leng Han
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Level 3 and 4, Singapore, 138648, Singapore
| | - Yingbin Liu
- Department of Biliary and Pancreatic Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youqiong Ye
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bing Su
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, and the Ministry of Education Key Laboratory of Cell Death and Differentiation, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Department of Gastroenterology, Center for Immune-related Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
195
|
Schirris Y, Gavves E, Nederlof I, Horlings HM, Teuwen J. DeepSMILE: Contrastive self-supervised pre-training benefits MSI and HRD classification directly from H&E whole-slide images in colorectal and breast cancer. Med Image Anal 2022; 79:102464. [DOI: 10.1016/j.media.2022.102464] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 03/21/2022] [Accepted: 04/15/2022] [Indexed: 02/07/2023]
|
196
|
Fang C, Zhong R, Qiu C, Zou BB. The Prognostic Value of GNG7 in Colorectal Cancer and Its Relationship With Immune Infiltration. Front Genet 2022; 13:833013. [PMID: 35281820 PMCID: PMC8906903 DOI: 10.3389/fgene.2022.833013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
Background: G Protein Subunit Gamma 7 (GNG7) is an important gene that regulates cell proliferation and induces apoptosis. However, the correlation between GNG7 expression and immune infiltration as well as patient prognosis of colorectal cancer (CRC) remains unclear. Methods: The GNG7 expression differences between tumor tissues and normal tissues were explored via the Oncomine database, Tumor Immune Estimation Resource (TIMER) site and UALCAN database. Then, the influence of GNG7 on clinical prognosis were evaluated, using the PrognoScan database. In addition, the relationship between GNG7 and tumor-related immune infiltration as well as gene marker sets of immune infiltration was investigated via TIMER, TISIDB and GEPIA. Results: We found that GNG7 expression was down-regulated in multiple malignant tumors including colorectal cancer (CRC) and the GNG7 expression was associated with tumor stage, histology subtype, lymph node metastasis and poor prognosis in colorectal cancer (CRC). In addition, the expression of GNG7 was significantly associated with infiltration level of multiple immune cells, immunomodulatory factors as well as part of the immune cell markers. Conclusion: GNG7 displays validated prognostic value in CRC and was associated with its immune cell infiltration and immunoregulation. These results suggest that GNG7 is a potential prognostic marker and is associated with tumor immune infiltration, thus providing a new perspective for the immunotherapy of CRC.
Collapse
Affiliation(s)
- Can Fang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rulei Zhong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chenyang Qiu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Bing-Bing Zou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
197
|
Bohaumilitzky L, Kluck K, Hüneburg R, Gallon R, Nattermann J, Kirchner M, Kristiansen G, Hommerding O, Pfuderer PL, Wagner L, Echterdiek F, Kösegi S, Müller N, Fischer K, Nelius N, Hartog B, Borthwick G, Busch E, Haag GM, Bläker H, Möslein G, von Knebel Doeberitz M, Seppälä TT, Ahtiainen M, Mecklin JP, Bishop DT, Burn J, Stenzinger A, Budczies J, Kloor M, Ahadova A. The Different Immune Profiles of Normal Colonic Mucosa in Cancer-Free Lynch Syndrome Carriers and Lynch Syndrome Colorectal Cancer Patients. Gastroenterology 2022; 162:907-919.e10. [PMID: 34863788 DOI: 10.1053/j.gastro.2021.11.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 11/02/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Owing to the high load of immunogenic frameshift neoantigens, tumors arising in individuals with Lynch syndrome (LS), the most common inherited colorectal cancer (CRC) syndrome, are characterized by a pronounced immune infiltration. However, the immune status of normal colorectal mucosa in LS is not well characterized. We assessed the immune infiltrate in tumor-distant normal colorectal mucosa from LS CRC patients, sporadic microsatellite-unstable (MSI) and microsatellite-stable (MSS) CRC patients, and cancer-free LS carriers. METHODS CD3-positive, FOXP3-positive, and CD8-positive T cells were quantified in, respectively, 219, 233, and 201 formalin-fixed paraffin-embedded (FFPE) normal colonic mucosa tissue sections from CRC patients and cancer-free LS carriers and 26, 22, and 19 LS CRCs. CD3-positive T cells were also quantified in an independent cohort of 97 FFPE normal rectal mucosa tissue sections from LS carriers enrolled in the CAPP2 clinical trial. The expression of 770 immune-relevant genes was analyzed in a subset of samples with the use of the NanoString nCounter platform. RESULTS LS normal mucosa specimens showed significantly elevated CD3-, FOXP3-, and CD8-positive T-cell densities compared with non-LS control specimens. Gene expression profiling and cluster analysis revealed distinct immune profiles in LS carrier mucosa with and without cancer manifestation. Long-term follow-up of LS carriers within the CAPP2 trial found a correlation between mucosal T-cell infiltrate and time to subsequent tumor occurrence. CONCLUSIONS LS carriers show elevated mucosal T-cell infiltration even in the absence of cancer. The normal mucosa immune profile may be a temporary or permanent tumor risk modifier in LS carriers.
Collapse
Affiliation(s)
- Lena Bohaumilitzky
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Robert Hüneburg
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany; National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Richard Gallon
- Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany; National Center for Hereditary Tumor Syndromes, University Hospital Bonn, Bonn, Germany
| | - Martina Kirchner
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | | | | | - Pauline L Pfuderer
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lelia Wagner
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Fabian Echterdiek
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Nephrology, Klinikum Stuttgart-Katharinenhospital, Stuttgart, Germany
| | - Svenja Kösegi
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nico Müller
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Konstantin Fischer
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nina Nelius
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ben Hartog
- Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Gillian Borthwick
- Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Elena Busch
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Georg Martin Haag
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Hendrik Bläker
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Gabriela Möslein
- Department of Surgery, Ev. Krankenhaus Bethesda Hospital, Duisburg, Germany
| | - Magnus von Knebel Doeberitz
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Toni T Seppälä
- Department of Gastrointestinal Surgery, Helsinki University Central Hospital, Helsinki, Finland; Applied Tumor Genomics Research Program, University of Helsinki, Helsinki, Finland
| | - Maarit Ahtiainen
- Department of Molecular Pathology, Central Finland Hospital Nova, Jyväskylä, Finland
| | - Jukka-Pekka Mecklin
- Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland; Department of Surgery, Central Finland Hospital Nova, Jyväskylä, Finland
| | - D Timothy Bishop
- Division of Haematology and Immunology, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - John Burn
- Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne, United Kingdom
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Budczies
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Aysel Ahadova
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany; Cooperation Unit Applied Tumor Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
198
|
Berle M, Hestetun KE, Vethe H, Chera S, Paulo JA, Dahl O, Myklebust MP. Mapping Proteome Changes in Microsatellite Stable, Recurrent Colon Cancer Reveals a Significant Immune System Signature. Cancer Genomics Proteomics 2022; 19:130-144. [PMID: 35181583 DOI: 10.21873/cgp.20309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND/AIM Better stratification of the risk of relapse will help select the right patients for adjuvant treatment and improve targeted therapies for patients with colon cancer. MATERIALS AND METHODS To understand why a subset of tumors relapse, we compared the proteome of two groups of patients with colon cancer with similar stage, stratified based on the presence or absence of recurrence. RESULTS Using tumor biopsies from the primary operation, we identified dissimilarity between recurrent and nonrecurrent mismatch satellite stable colon cancer and found that signaling related to immune activation and inflammation was associated with relapse. CONCLUSION Immune modulation may have an effect on mismatch satellite stable colon cancer. At present, immune therapy is offered primarily to microsatellite instable colon cancer. Hopefully, immune therapy in mismatch satellite stable colon cancer beyond PD-1 and PD-L1 inhibitors can be implemented.
Collapse
Affiliation(s)
- Magnus Berle
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; .,Department of Surgery, Haukeland University Hospital, Bergen, Norway.,Department of Surgery, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Kjersti E Hestetun
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | - Heidrun Vethe
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Simona Chera
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Medicine, Division of Endocrinology, Diabetes, Nutrition and Patient Education, University Hospital of Geneva, Geneva, Switzerland
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, U.S.A
| | - Olav Dahl
- Department of Clinical Science, University of Bergen, Bergen, Norway.,Department of Oncology and Medical Physics, Haukeland University Hospital, Bergen, Norway
| | | |
Collapse
|
199
|
Bai Z, Zhou Y, Ye Z, Xiong J, Lan H, Wang F. Tumor-Infiltrating Lymphocytes in Colorectal Cancer: The Fundamental Indication and Application on Immunotherapy. Front Immunol 2022; 12:808964. [PMID: 35095898 PMCID: PMC8795622 DOI: 10.3389/fimmu.2021.808964] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/24/2021] [Indexed: 12/22/2022] Open
Abstract
The clinical success of immunotherapy has revolutionized the treatment of cancer patients, bringing renewed attention to tumor-infiltrating lymphocytes (TILs) of various cancer types. Immune checkpoint blockade is effective in patients with mismatched repair defects and high microsatellite instability (dMMR-MSI-H) in metastatic colorectal cancer (CRC), leading the FDA to accelerate the approval of two programmed cell death 1 (PD-1) blocking antibodies, pembrolizumab and nivolumab, for treatment of dMMR-MSI-H cancers. In contrast, patients with proficient mismatch repair and low levels of microsatellite stability or microsatellite instability (pMMR-MSI-L/MSS) typically have low tumor-infiltrating lymphocytes and have shown unsatisfied responses to the immune checkpoint inhibitor. Different TILs environments reflect different responses to immunotherapy, highlighting the complexity of the underlying tumor-immune interaction. Profiling of TILs fundamental Indication would shed light on the mechanisms of cancer-immune evasion, thus providing opportunities for the development of novel therapeutic strategies. In this review, we summarize phenotypic diversities of TILs and their connections with prognosis in CRC and provide insights into the subsets-specific nature of TILs with different MSI status. We also discuss current clinical immunotherapy approaches based on TILs as well as promising directions for future expansion, and highlight existing clinical data supporting its use.
Collapse
Affiliation(s)
- Ziyi Bai
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China.,College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yao Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zifan Ye
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jialong Xiong
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Hongying Lan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Feng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
200
|
Elsayed I, Elsayed N, Feng Q, Sheahan K, Moran B, Wang X. Multi-OMICs data analysis identifies molecular features correlating with tumor immunity in colon cancer. Cancer Biomark 2022; 33:261-271. [PMID: 35213358 DOI: 10.3233/cbm-210222] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND There is a current need for new markers with higher sensitivity and specificity to predict immune status and optimize immunotherapy use in colon cancer. OBJECTIVE We aimed to investigate the multi-OMICs features associated with colon cancer immunity and response to immunotherapy. METHODS We evaluated the association of multi-OMICs data from three colon cancer datasets (TCGA, CPTAC2, and Samstein) with antitumor immune signatures (CD8+ T cell infiltration, immune cytolytic activity, and PD-L1 expression). Using the log-rank test and hierarchical clustering, we explored the association of various OMICs features with survival and immune status in colon cancer. RESULTS Two gene mutations (TERT and ERBB4) correlated with antitumor cytolytic activity found also correlated with improved survival in immunotherapy-treated colon cancers. Moreover, the expression of numerous genes was associated with antitumor immunity, including GBP1, GBP4, GBP5, NKG7, APOL3, IDO1, CCL5, and CXCL9. We clustered colon cancer samples into four immuno-distinct clusters based on the expression levels of 82 genes. We have also identified two proteins (PREX1 and RAD50), ten miRNAs (hsa-miR-140, 146, 150, 155, 342, 59, 342, 511, 592 and 1977), and five oncogenic pathways (CYCLIN, BCAT, CAMP, RB, NRL, EIF4E, and VEGF signaling pathways) significantly correlated with antitumor immune signatures. CONCLUSION These molecular features are potential markers of tumor immune status and response to immunotherapy.
Collapse
Affiliation(s)
- Inas Elsayed
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, Jiangsu, China.,Department of Pharmacology, Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | - Nazik Elsayed
- Department of Statistics, Faculty of Mathematics and Computer Sciences, University of Gezira, Wad Madani, Sudan
| | - Qiushi Feng
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Kieran Sheahan
- Centre for Colorectal Disease, St. Vincent's University Hospital, Elm Park, Ireland.,School of Medicine and Medical Sciences, University College Dublin, Belfield, Ireland
| | - Bruce Moran
- Department of Pathology, St. Vincent's University Hospital, Elm Park, Ireland
| | - Xiaosheng Wang
- Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.,Big Data Research Institute, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|