1
|
Abbott AG, Meyers DE, Elmi-Assadzadeh G, Stukalin I, Marro A, Puloski SKT, Morris DG, Cheung WY, Monument MJ. Effectiveness of immune checkpoint inhibitor therapy on bone metastases in non-small-cell lung cancer. Front Immunol 2024; 15:1379056. [PMID: 38957472 PMCID: PMC11217176 DOI: 10.3389/fimmu.2024.1379056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Background Bone metastases (BoMs) are prevalent in patients with metastatic non-small-cell lung cancer (NSCLC) however, there are limited data detailing how BoMs respond to immune checkpoint inhibitors (ICIs). The purpose of this study was to compare the imaging response to ICIs of BoMs against visceral metastases and to evaluate the effect of BoMs on survival. Materials and methods A retrospective, multicentre cohort study was conducted in patients with NSCLC treated with nivolumab or pembrolizumab in Alberta, Canada from 2015 to 2020. The primary endpoint was the real-world organ specific progression free survival (osPFS) of bone versus visceral metastases. Visceral metastases were categorized as adrenal, brain, liver, lung, lymph node, or other intra-abdominal lesions. The secondary outcome was overall survival (OS) amongst patients with and without BoMs. Results A total of 573 patients were included of which all patients had visceral metastases and 243 patients (42.4%) had BoMs. High PD-L1 expression was identified in 268 patients (46.8%). No significant difference in osPFS was observed between bone, liver, and intra-abdominal metastases (p=0.20 and p=0.76, respectively), with all showing shorter osPFS than other disease sites. There was no difference in the osPFS of extra-thoracic sites of disease in patients with high PD-L1 expression. There was significant discordance between visceral disease response and bone disease response to ICI (p=0.047). The presence of BoMs was an independent poor prognostic factor for OS (HR 1.26, 95%CI: 1.05-1.53, p=0.01). Conclusion Metastatic bone, liver, and intra-abdominal lesions demonstrated inferior clinical responses to ICI relative to other sites of disease. Additionally, the presence of bone and liver metastases were independent poor prognostic factors for overall survival. This real-world data suggests that BoMs respond poorly to ICI and may require treatment adjuncts for disease control.
Collapse
Affiliation(s)
- Annalise G. Abbott
- Section of Orthopaedic Surgery, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Daniel E. Meyers
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | | | - Igor Stukalin
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Alessandro Marro
- Departmenmt of Radiology, University of Calgary, Calgary, AB, Canada
| | - Shannon K. T. Puloski
- Section of Orthopaedic Surgery, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone & Joint Institute, University of Calgary, Calgary, AB, Canada
| | - Don G. Morris
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Winson Y. Cheung
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Michael J. Monument
- Section of Orthopaedic Surgery, University of Calgary, Calgary, AB, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
- McCaig Bone & Joint Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
2
|
Wen Q, Wang S, Hong L, Shen S, He Y, Sheng X, Zhuang X, Chen S, Wang Y, Zhuang H. m 5 C regulator-mediated methylation modification patterns and tumor microenvironment infiltration characteristics in acute myeloid leukemia. Immun Inflamm Dis 2024; 12:e1150. [PMID: 38270308 PMCID: PMC10802208 DOI: 10.1002/iid3.1150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/11/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Recently, many studies have been conducted to examine immune response modification at epigenetic level, but the candidate effect of RNA 5-methylcytosine (m5 C) modification on tumor microenvironment (TME) of acute myeloid leukemia (AML) is still unknown at present. METHODS We assessed the patterns of m5 C modification among 417 AML cases by using nine m5 C regulators. Thereafter, we associated those identified modification patterns with TME cell infiltration features. Additionally, stepwise regression and LASSO Cox regression analyses were conducted for quantifying patterns of m5 C modification among AML cases to establish the m5 C-score. Meanwhile, we validated the expression of genes in the m5C-score model by qRT-PCR. Finally, the present work analyzed the association between m5 C-score and AML clinical characteristics and prognostic outcomes. RESULTS In total, three different patterns of m5 C modification (m5 C-clusters) were identified, and highly differentiated TME cell infiltration features were also identified. On this basis, evaluating patterns of m5 C modification in single cancer samples was important for evaluating the immune/stromal activities in TME and for predicting prognosis. In addition, the m5 C-score was established, which showed a close relation with the overall survival (OS) of test and training set samples. Moreover, multivariate Cox analysis suggested that our constructed m5 C-score served as the independent predicting factor for the prognosis of AML (hazard ratio = 1.57, 95% confidence interval = 1.38-1.79, p < 1e-5 ). CONCLUSIONS This study shows that m5 C modification may be one of the key roles in the formation of diversity and complexity of TME. Meanwhile, assessing the patterns of m5 C modification among individual cancer samples is of great importance, which provides insights into cell infiltration features within TME, thereby helping to develop relevant immunotherapy and predict patient prognostic outcomes.
Collapse
Affiliation(s)
- Qiang Wen
- Department of Gynecologic OncologyCancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)HangzhouZhejiangChina
| | - ShouJun Wang
- Department of MedicineHangZhou FuYang Hospital of Traditional Chinese MedicineHangzhouZhejiangChina
| | - Lili Hong
- Department of Hematology and TransfusionThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Hang ZhouZhejiangChina
| | - Siyu Shen
- The First School of Clinical MedicineZhejiang Chinese Medical UniversityHangzhouZhejiangChina
| | - Yibo He
- Department of Clinical LabThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouZhejiangChina
| | - Xianfu Sheng
- Department of Hematology and TransfusionThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Hang ZhouZhejiangChina
| | - Xiaofen Zhuang
- Department of MedicineHangZhou FuYang Hospital of Traditional Chinese MedicineHangzhouZhejiangChina
| | - Shiliang Chen
- Department of Clinical LabThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)HangzhouZhejiangChina
| | - Ying Wang
- Department of Clinical Research Center, Affiliated Hangzhou First People's HospitalZhejiang University School of MedicineHangzhouZhejiangChina
| | - Haifeng Zhuang
- Department of Hematology and TransfusionThe First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine)Hang ZhouZhejiangChina
| |
Collapse
|
3
|
Deng JY, Gou Q, Yang L, Chen ZH, Yang MY, Yang XR, Yan HH, Wei XW, Liu JQ, Su J, Zhong WZ, Xu CR, Wu YL, Zhou Q. Immune suppressive microenvironment in liver metastases contributes to organ-specific response of immunotherapy in advanced non-small cell lung cancer. J Immunother Cancer 2023; 11:e007218. [PMID: 37463790 PMCID: PMC10357800 DOI: 10.1136/jitc-2023-007218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND The liver is a frequent site of metastases and liver metastases (LM) correlate with diminished immunotherapy efficacy in non-small cell lung cancer (NSCLC). This study aimed to analyze whether tumor response to immunotherapy differs between pulmonary lesions (PL) and LM in NSCLC and to explore potential mechanisms through multiomics analysis. METHODS This observational longitudinal clinical cohort study included patients with NSCLC with LM receiving immunotherapy was conducted to evaluate organ-specific tumor response of PL and LM. We collected paired PL and LM tumor samples to analyze the organ-specific difference using whole-exome sequencing, RNA sequencing, and multiplex immunohistochemistry. RESULTS A total of 52 patients with NSCLC with LM were enrolled to evaluate the organ-specific response of immunotherapy. The objective response rate (21.1% vs 32.7%) and disease control rate of LM were lower than that of PL (67.3% vs 86.5%). One-third of patients showed mixed response, among whom 88.2% (15/17) presented with LM increasing, but PL decreasing, while the others had the opposite pattern (p=0.002). In another independent cohort, 27 pairs of matched PL and LM tumor samples from the same individuals, including six simultaneously collected pairs, were included in the translational part. Genomic landscapes profiling revealed similar somatic mutations, tumor mutational burden, and neoantigen number between PL and LM. Bulk-RNA sequencing showed immune activation-related genes including CD8A, LCK, and ICOS were downregulated in LM. The antigen processing and presentation, natural killer (NK) cell-mediated cytotoxicity and T-cell receptor signaling pathway were enriched in PL compared with LM. Multiplex immunohistochemistry detected significantly lower fractions of CD8+ cells (p=0.036) and CD56dim+ cells (p=0.016) in LM compared with PL. Single-cell RNA sequencing also characterized lower effector CD8+ T cells activation and NK cells cytotoxicity in LM. CONCLUSIONS Compared with PL, LM presents an inferior organ-specific tumor response to immunotherapy. PL and LM showed limited heterogeneity in the genomic landscape, while the LM tumor microenvironment displayed lower levels of immune activation and infiltration than PL, which might contribute to developing precise immunotherapy strategies for patients with NSCLC with LM.
Collapse
Affiliation(s)
- Jia-Yi Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Qing Gou
- Department of Interventional Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lingling Yang
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc, Nanjing, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ming-Yi Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Xiao-Rong Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xue-Wu Wei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jia-Qi Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
4
|
Yuan C, Zhao X, Wangmo D, Alshareef D, Gates TJ, Subramanian S. Tumor models to assess immune response and tumor-microbiome interactions in colorectal cancer. Pharmacol Ther 2022; 231:107981. [PMID: 34480964 PMCID: PMC8844062 DOI: 10.1016/j.pharmthera.2021.107981] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/20/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023]
Abstract
Despite significant advances over the past 2 decades in preventive screening and therapy aimed at improving patient survival, colorectal cancer (CRC) remains the second most common cause of cancer death in the United States. The average 5-year survival rate of CRC patients with positive regional lymph nodes is only 40%, while less than 5% of patients with distant metastases survive beyond 5 years. There is a critical need to develop novel therapies that can improve overall survival in patients with poor prognoses, particularly since 60% of them are diagnosed at an advanced stage. Pertinently, immune checkpoint blockade therapy has dramatically changed how we treat CRC patients with microsatellite-instable high tumors. Furthermore, accumulating evidence shows that changes in gut microbiota are associated with the regulation of host antitumor immune response and cancer progression. Appropriate animal models are essential to deciphering the complex mechanisms of host antitumor immune response and tumor-gut microbiome metabolic interactions. Here, we discuss various mouse models of colorectal cancer that are developed to address key questions on tumor immune response and tumor-microbiota interactions. These CRC models will also serve as resourceful tools for effective preclinical studies.
Collapse
Affiliation(s)
- Ce Yuan
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Xianda Zhao
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Dechen Wangmo
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Duha Alshareef
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Travis J Gates
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Subbaya Subramanian
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, United States of America; Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States of America; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, United States of America.
| |
Collapse
|
5
|
Jiménez-Chávez ÁDJ, Nava-García BK, Bustos-Jaimes I, Moreno-Fierros L. B19-VLPs as an effective delivery system for tumour antigens to induce humoral and cellular immune responses against triple negative breast cancer. Immunol Lett 2021; 239:77-87. [PMID: 34508790 DOI: 10.1016/j.imlet.2021.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/25/2021] [Accepted: 09/03/2021] [Indexed: 01/16/2023]
Abstract
Cancer immunotherapy is emerging as a viable treatment option for several types of cancer. Active immunotherapy aims for the induction of specific antitumor immune responses; this goal requires strategies capable of increasing the immunogenicity of tumour antigens. Parvovirus B19 virus-like particles (B19-VLPs) formed of VP2 protein had been shown to be an effective multi-neoepitope delivery system capable of inducing specific cellular responses towards coupled antigens and reducing tumour growth and lung metastases in triple negative breast cancer mouse model. These findings encouraged us to further characterise these VP2 B19-VLPs by testing their capacity to simultaneously induce cellular and humoral responses towards other tumour-associated antigens, as this had not yet been evaluated. Here, we designed and evaluated in the 4T1 breast cancer model the prophylactic and therapeutic effect of VP2 B19-VLPs decorated with cellular (P53) and humoral (MUC1) epitopes. Balb/c mice were immunised with chimaeric VLPs, vehicle, or VLPs plus adjuvant. Tumour establishment and growth, lung metastasis, and cellular and humoral immune responses were evaluated. The prophylactic administration of chimaeric VLPs without adjuvant prevented the establishment of the tumour, while by therapeutic administration, chimaeric VLPs induced smaller tumour growth and decreased the number of metastases in the lung compared to wild-type VLPs. chimaeric VLPs induced high antibody titres towards the MUC1 epitope, as well as specific cellular responses towards P53 epitopes in lymph nodes local to the tumour. Our results reinforce and extend the utility of VP2 B19-VLPs as an encouraging tumour antigen delivery system in cancer immunotherapy able to improve tumour immunity in TNBC by inducing cellular and humoral immune responses.
Collapse
Affiliation(s)
- Ángel de Jesús Jiménez-Chávez
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Brenda Katherine Nava-García
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México
| | - Ismael Bustos-Jaimes
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
| | - Leticia Moreno-Fierros
- Biomedicine Research Unit, Faculty of Higher Studies Iztacala, National Autonomous University of Mexico. Avenida de los Barrios 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, 54090, México.
| |
Collapse
|
6
|
Ho WJ, Erbe R, Danilova L, Phyo Z, Bigelow E, Stein-O'Brien G, Thomas DL, Charmsaz S, Gross N, Woolman S, Cruz K, Munday RM, Zaidi N, Armstrong TD, Sztein MB, Yarchoan M, Thompson ED, Jaffee EM, Fertig EJ. Multi-omic profiling of lung and liver tumor microenvironments of metastatic pancreatic cancer reveals site-specific immune regulatory pathways. Genome Biol 2021; 22:154. [PMID: 33985562 PMCID: PMC8118107 DOI: 10.1186/s13059-021-02363-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The majority of pancreatic ductal adenocarcinomas (PDAC) are diagnosed at the metastatic stage, and standard therapies have limited activity with a dismal 5-year survival rate of only 8%. The liver and lung are the most common sites of PDAC metastasis, and each have been differentially associated with prognoses and responses to systemic therapies. A deeper understanding of the molecular and cellular landscape within the tumor microenvironment (TME) metastasis at these different sites is critical to informing future therapeutic strategies against metastatic PDAC. RESULTS By leveraging combined mass cytometry, immunohistochemistry, and RNA sequencing, we identify key regulatory pathways that distinguish the liver and lung TMEs in a preclinical mouse model of metastatic PDAC. We demonstrate that the lung TME generally exhibits higher levels of immune infiltration, immune activation, and pro-immune signaling pathways, whereas multiple immune-suppressive pathways are emphasized in the liver TME. We then perform further validation of these preclinical findings in paired human lung and liver metastatic samples using immunohistochemistry from PDAC rapid autopsy specimens. Finally, in silico validation with transfer learning between our mouse model and TCGA datasets further demonstrates that many of the site-associated features are detectable even in the context of different primary tumors. CONCLUSIONS Determining the distinctive immune-suppressive features in multiple liver and lung TME datasets provides further insight into the tissue specificity of molecular and cellular pathways, suggesting a potential mechanism underlying the discordant clinical responses that are often observed in metastatic diseases.
Collapse
Affiliation(s)
- Won Jin Ho
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- The Johns Hopkins Cancer Convergence Institute, Baltimore, USA
- Skip Viragh Center for Pancreatic Cancer, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 4M07 Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Rossin Erbe
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, USA
| | - Ludmila Danilova
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Zaw Phyo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Emma Bigelow
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | | | - Dwayne L Thomas
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Soren Charmsaz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Nicole Gross
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Skylar Woolman
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Kayla Cruz
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Rebecca M Munday
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, USA
| | - Neeha Zaidi
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- Skip Viragh Center for Pancreatic Cancer, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 4M07 Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - Todd D Armstrong
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
| | - Elizabeth D Thompson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA
- Skip Viragh Center for Pancreatic Cancer, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 4M07 Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD, 21287, USA
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, USA
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA.
- The Johns Hopkins Cancer Convergence Institute, Baltimore, USA.
- Skip Viragh Center for Pancreatic Cancer, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 4M07 Bunting Blaustein Cancer Research Building, 1650 Orleans Street, Baltimore, MD, 21287, USA.
| | - Elana J Fertig
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 550 N Broadway Suite 1101E, Baltimore, MD, 21209, USA.
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, USA.
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, USA.
| |
Collapse
|
7
|
Co-inhibitor expression on tumor infiltrating and splenic lymphocytes after dual checkpoint inhibition in a microsatellite stable model of colorectal cancer. Sci Rep 2021; 11:6956. [PMID: 33772035 PMCID: PMC7997991 DOI: 10.1038/s41598-021-85810-5] [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: 08/20/2020] [Accepted: 02/18/2021] [Indexed: 01/08/2023] Open
Abstract
Checkpoint inhibitors have demonstrated clinical impact in colorectal cancer with deficient mismatch repair and high microsatellite instability. However, the majority of patients have disease with stable microsatellites that responds poorly to immunotherapies. Combinations of checkpoint inhibitors are under investigation as a way of increasing immunogenicity and promoting a robust anti-tumor immune response. The purpose of this study is to quantify the immune responses induced by mono and dual checkpoint inhibition in a mismatch repair proficient model of colorectal cancer (CRC). Tumor growth rates were monitored over time and compared between groups. We utilized fluorescence-activated cell sorting to analyze CD8+ and CD4+ T cells after treatment with either single PD-1 inhibition or dual PD-1 and CTLA-4 inhibition. Additionally, we sought to quantify the expression of co-inhibitory surface molecules PD-1, LAG3, and TIM3. Dual checkpoint inhibition was associated with a significantly slower growth rate as compared to either mono PD-1 inhibition or control (p < 0.05). Neither monotherapy nor dual checkpoint inhibition significantly affected the tumoral infiltration of lymphocytes. After treatment with dual inhibitors, infiltrating CD8+ T cells demonstrated significantly less expression of PD-1 (1700 vs. 2545 and 2462; p < 0.05) and LAG3 (446.2 vs. 694.4 and 707; p < 0.05) along with significantly more expression of TIM3 (12,611 vs. 2961 and 4259; p < 0.05) versus the control and anti-PD-1 groups. These results suggest that dual therapy with anti-CTLA-4 and anti-PD-1 antibodies significantly inhibits growth of microsatellite stable CRC by suppressing immunosuppressive checkpoints. Upregulation of TIM3 represents a potential escape mechanism and a target for future combination immunotherapies in CRC.
Collapse
|
8
|
Oliver AJ, Davey AS, Keam SP, Mardiana S, Chan JD, von Scheidt B, Beavis PA, House IG, Van Audernaerde JR, Darcy PK, Kershaw MH, Slaney CY. Tissue-specific tumor microenvironments influence responses to immunotherapies. Clin Transl Immunology 2019; 8:e1094. [PMID: 31768254 PMCID: PMC6869967 DOI: 10.1002/cti2.1094] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/24/2019] [Accepted: 10/30/2019] [Indexed: 01/18/2023] Open
Abstract
Objectives Investigation of variable response rates to cancer immunotherapies has exposed the immunosuppressive tumor microenvironment (TME) as a limiting factor of therapeutic efficacy. A determinant of TME composition is the tumor location, and clinical data have revealed associations between certain metastatic sites and reduced responses. Preclinical models to study tissue‐specific TMEs have eliminated genetic heterogeneity, but have investigated models with limited clinical relevance. Methods We investigated the TMEs of tumors at clinically relevant sites of metastasis (liver and lungs) and their impact on αPD‐1/αCTLA4 and trimAb (αDR5, α4‐1BB, αCD40) therapy responses in the 67NR mouse breast cancer and Renca mouse kidney cancer models. Results Tumors grown in the lungs were resistant to both therapies whereas the same tumor lines growing in the mammary fat pad (MFP), liver or subcutaneously could be completely eradicated, despite greater tumor burden. Assessment of tumor cells and drug delivery in 67NR lung or MFP tumors revealed no differences and prompted investigation into the immune TME. Lung tumors had a more immunosuppressive TME with increased myeloid‐derived suppressor cell infiltration, decreased T cell infiltration and activation, and decreased NK cell activation. Depletion of various immune cell subsets indicated an equivalent role for NK cells and CD8+ T cells in lung tumour control. Thus, targeting T cells with αPD‐1/αCTLA4 or trimAb was not sufficient to elicit a robust antitumor response in lung tumors. Conclusion Taken together, these data demonstrate that tissue‐specific TMEs influence immunotherapy responses and highlight the importance in defining tissue‐specific response patterns in patients.
Collapse
Affiliation(s)
- Amanda J Oliver
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Ashleigh S Davey
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Simon P Keam
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Tumour Suppression Laboratory Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Sherly Mardiana
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Jack D Chan
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Bianca von Scheidt
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia
| | - Paul A Beavis
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Imran G House
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Jonas Rm Van Audernaerde
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Center for Oncological Research Faculty of Medicine and Health Sciences University of Antwerp Antwerp Belgium
| | - Phillip K Darcy
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Michael H Kershaw
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| | - Clare Y Slaney
- Cancer Immunology Program Peter MacCallum Cancer Centre Melbourne VIC Australia.,Sir Peter MacCallum Department of Oncology The University of Melbourne Parkville VIC Australia
| |
Collapse
|
9
|
Zhang C, Yu D. Suppressing immunotherapy by organ-specific tumor microenvironments: what is in the brain? Cell Biosci 2019; 9:82. [PMID: 31624532 PMCID: PMC6781341 DOI: 10.1186/s13578-019-0349-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022] Open
Abstract
Recent breakthroughs in cancer immunotherapy have led to curative efficacy and significantly prolonged survival in a subset of patients of multiple cancer types; and immunotherapy has become the newest pillar of cancer treatment in addition to surgery, chemotherapy, radiotherapy and precision targeted therapies. In the metastatic disease setting, responses to immunotherapy are heterogeneous depending on the metastatic organ sites. The tissue-specific immuno-biology in the tumor microenvironments (TMEs) contributes to the differential therapeutic responses. Herein, we review the impact of tissue-specific tumor microenvironment on the efficacy of immunotherapy, with a focus on historically under-represented central nervous system (CNS) metastasis, which was excluded from most clinical trials. Retrospective examination of patient specimens and prospective clinical studies with immune checkpoint blockade (ICB) have established that brain can harbor an “active” immune microenvironment for effective immunotherapy. Regulation by the innate immune microglial cells and remodeling of the blood–brain barrier (BBB) may contribute to immunotherapeutic responses mediated by T lymphocytes. How to convert an “inactive” (cold) brain microenvironment into an “active” (hot) brain TME should be the focus of future efforts. Thus, procurement and complete examination of clinical specimens from brain metastases as well as development of appropriate preclinical brain metastasis models susceptible to external manipulation of the TME are critical steps towards that goal. A deeper understanding of the immuno-biology in distinct organ microenvironments will help to expand the benefits of immunotherapy to more needed patients.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
10
|
Characterization of immune cell subtypes in three commonly used mouse strains reveals gender and strain-specific variations. J Transl Med 2019; 99:93-106. [PMID: 30353130 PMCID: PMC6524955 DOI: 10.1038/s41374-018-0137-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/22/2018] [Accepted: 09/12/2018] [Indexed: 12/21/2022] Open
Abstract
The lack of consensus on bone marrow (BM) and splenic immune cell profiles in preclinical mouse strains complicates comparative analysis across different studies. Although studies have documented relative distribution of immune cells from peripheral blood in mice, similar studies for BM and spleen from naïve mice are lacking. In an effort to establish strain- and gender-specific benchmarks for distribution of various immune cell subtypes in these organs, we performed immunophenotypic analysis of BM cells and splenocytes from both genders of three commonly used murine strains (C57BL/6NCr, 129/SvHsd, and BALB/cAnNCr). Total neutrophils and splenic macrophages were significantly higher in C57BL/6NCr, whereas total B cells were lower. Within C57BL/6NCr female mice, BM B cells were elevated with respect to the males whereas splenic mDCs and splenic neutrophils were reduced. Within BALB/cAnNCr male mice, BM CD4+ Tregs were elevated with respect to the other strains. Furthermore, in male BALB/cAnNCr mice, NK cells were elevated with respect to the other strains in both BM and spleen. Splenic CD4+ Tregs and splenic CD8+ T cells were reduced in male BALB/c mice in comparison to female mice. Bone marrow CD4+ T cells and mDCs were significantly increased in 129/SvHsd whereas splenic CD8+ T cells were reduced. In general, males exhibited higher immature myeloid cells, macrophages, and NK cells. To our knowledge, this study provides a first attempt to systematically establish organ-specific benchmarks on immune cells in studies involving these mouse strains.
Collapse
|
11
|
Nanofluidic drug-eluting seed for sustained intratumoral immunotherapy in triple negative breast cancer. J Control Release 2018; 285:23-34. [DOI: 10.1016/j.jconrel.2018.06.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/15/2018] [Accepted: 06/28/2018] [Indexed: 12/11/2022]
|
12
|
ADAMTS1 protease is required for a balanced immune cell repertoire and tumour inflammatory response. Sci Rep 2018; 8:13103. [PMID: 30166561 PMCID: PMC6117274 DOI: 10.1038/s41598-018-31288-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 08/16/2018] [Indexed: 12/21/2022] Open
Abstract
Recent advances have emphasized the relevance of studying the extracellular microenvironment given its main contribution to tissue homeostasis and disease. Within this complex scenario, we have studied the extracellular protease ADAMTS1 (a disintegrin and metalloprotease with thrombospondin motif 1), implicated in vascularization and development, with reported anti- and pro-tumorigenic activities. In this work we performed a detailed study of the vasculature and substrates in adult organs of wild type and Adamts1-deficient mice. In addition to the expected alterations of organs like kidney, heart and aorta, we found that the lack of ADAMTS1 differently affects lymphocyte and myeloid populations in the spleen and bone marrow. The study of the substrate versican also revealed its alteration in the absence of the protease. With such premises, we challenged our mice with subcutaneous B16F1 syngeneic tumours and closely evaluated the immune repertoire in the tumours but also in the distant spleen and bone marrow. Our results confirmed a pro-inflammatory landscape in the absence of ADAMTS1, correlating with tumour blockade, supporting its novel role as a modulator of the immune cell response.
Collapse
|
13
|
Dorta-Estremera S, Chin RL, Sierra G, Nicholas C, Yanamandra AV, Nookala SMK, Yang G, Singh S, Curran MA, Sastry KJ. Mucosal HPV E6/E7 Peptide Vaccination in Combination with Immune Checkpoint Modulation Induces Regression of HPV + Oral Cancers. Cancer Res 2018; 78:5327-5339. [PMID: 30054333 DOI: 10.1158/0008-5472.can-18-0892] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/15/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022]
Abstract
High-risk human papillomavirus (HPV)-associated squamous cell carcinomas of the oropharynx (SCCOP) are among the fastest growing cancers. After standard-of-care treatment, however, patients with HPV+ SCCOP have better overall and disease-specific survival than patients with HPV- SCCOP, suggesting the importance of HPV-specific immunity. We reasoned that therapeutic vaccination targeting the HPV-16 E6 and E7 oncogenes could elicit high-affinity, high-frequency tumor antigen-specific T-cell responses, which could then be augmented and shielded from suppression in the tumor microenvironment by immune checkpoint modulation. In this study, we used a preclinical syngeneic mouse model of oral cancer comprised of mouse tonsil-derived epithelial cells stably expressing HPV-16 E6 and E7 genes along with H-ras oncogene (mEER) to identify combinations of vaccination and checkpoint antibodies capable of promoting tumor regression. Intranasal HPV E6/E7 peptide vaccination and single checkpoint antibodies failed to elicit responses in more than half of animals; however, 4-1BB agonist antibody along with either CD40 agonist antibody or CTLA-4 blockade eliminated the majority of established mEER tumors. The combination of intranasal HPV peptide vaccine and α4-1BB and αCTLA-4 antibodies produced curative efficacy and a better safety profile against orally implanted mEER tumors. Correlates of protective immunity included enhanced intratumoral levels of CD8 T cells relative to immunosuppressive regulatory T cells and myeloid-derived suppressor cells. Overall, our results demonstrate combination vaccine-immunotherapy modalities as novel treatment options for HPV+ SCCOP.Significance: Combinations of vaccine and checkpoint modulation are effective and safe treatment options for HPV+ oral cancers. Cancer Res; 78(18); 5327-39. ©2018 AACR.
Collapse
Affiliation(s)
| | - Renee L Chin
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gloria Sierra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - Courtney Nicholas
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ananta V Yanamandra
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sita M K Nookala
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Guojun Yang
- The University of Texas MD Anderson Cancer Center, Oncology Research for Biologics and Immunotherapy Translation, Houston, Texas, Texas
| | - Shail Singh
- Department of Melanoma and Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, Texas
| | - Michael A Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| | - K Jagannadha Sastry
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences at Houston, Houston, Texas
| |
Collapse
|
14
|
Oliver AJ, Lau PKH, Unsworth AS, Loi S, Darcy PK, Kershaw MH, Slaney CY. Tissue-Dependent Tumor Microenvironments and Their Impact on Immunotherapy Responses. Front Immunol 2018; 9:70. [PMID: 29445373 PMCID: PMC5797771 DOI: 10.3389/fimmu.2018.00070] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 01/10/2018] [Indexed: 12/11/2022] Open
Abstract
Recent advances in cancer immunology have led to a better understanding of the role of the tumor microenvironment (TME) in tumor initiation, progression, and metastasis. Tumors can occur at many locations within the body and coevolution between malignant tumor cells and non-malignant cells sculpts the TME at these sites. It has become increasingly clear that there are specific differences of the TMEs at different anatomical locations, and these tissue-specific TMEs regulate tumor growth, determine metastatic progression, and impact on the outcome of therapy responses. Herein, we review the scientific advances in understanding tissue-specific TMEs, discuss their impact on immunotherapeutic response, and assess the current clinical knowledge in this emerging field. A deeper understanding of the tissue-specific TME will help to develop effective immunotherapies against tumors and their metastases and assist in predicting clinical outcomes.
Collapse
Affiliation(s)
- Amanda J Oliver
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Peter K H Lau
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Ashleigh S Unsworth
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Sherene Loi
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Michael H Kershaw
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Clare Y Slaney
- Cancer Immunology Program, Peter MacCallum Cancer Center, Melbourne, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|