1
|
Haddad AF, Young JS, Gill S, Aghi MK. Resistance to immune checkpoint blockade: Mechanisms, counter-acting approaches, and future directions. Semin Cancer Biol 2022; 86:532-541. [PMID: 35276342 PMCID: PMC9458771 DOI: 10.1016/j.semcancer.2022.02.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 01/27/2023]
Abstract
Immunotherapies seek to unleash the immune system against cancer cells. While a variety of immunotherapies exist, one of the most commonly used is immune checkpoint blockade, which refers to the use of antibodies to interfere with immunosuppressive signaling through immune checkpoint molecules. Therapies against various checkpoints have had success in the clinic across cancer types. However, the efficacy of checkpoint inhibitors has varied across different cancer types and non-responsive patient populations have emerged. Non-responders to these therapies have highlighted the importance of understanding underlying mechanisms of resistance in order to predict which patients will respond and to tailor individual treatment paradigms. In this review we discuss the literature surrounding tumor mediated mechanisms of immune checkpoint resistance. We also describe efforts to overcome resistance and combine checkpoint inhibitors with additional immunotherapies. Finally, we provide insight into the future of immune checkpoint blockade, including the need for improved preclinical modeling and predictive biomarkers to facilitate personalized cancer treatments for patients.
Collapse
Affiliation(s)
| | | | | | - Manish K. Aghi
- Corresponding author at: Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Ave, M-779, San Francisco, CA 94143-0112, USA. (M.K. Aghi)
| |
Collapse
|
2
|
Tokaz MC, Baik CS, Houghton AM, Tseng D. New Immuno-oncology Targets and Resistance Mechanisms. Curr Treat Options Oncol 2022; 23:1201-1218. [PMID: 35980521 DOI: 10.1007/s11864-022-01005-8] [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] [Accepted: 07/27/2022] [Indexed: 11/28/2022]
Abstract
OPINION STATEMENT Immune checkpoint inhibition (ICI) has revolutionized the field of non-small cell lung cancer (NSCLC); currently, most patients with advanced disease receive upfront ICI either alone or in combination with chemotherapy. These advances have recently extended into early-stage NSCLC, with ICI incorporation into neoadjuvant and adjuvant treatment regimens. However, despite these successes, immunotherapy (IO) resistance remains a fundamental challenge in NSCLC, introducing a central quandary of how to precisely select the appropriate IO therapy or IO combination therapy for each individual patient. To address this vital need in the field, there has been an explosion of research in immuno-oncology to identify mechanisms of resistance, ranging from genomic alterations in the tumor to immunosuppressive conditions in the tumor microenvironment (TME). There remain many questions about how this complex interplay between the tumor and the immune microenvironment translates into clinical phenotypes of primary and acquired resistance. In NSCLC, a number of novel therapeutics are being developed to prevent and overcome resistance to ICI. Particular promise has been shown with therapeutics targeting novel T cell immune checkpoint inhibitors and targeting innate immune cells in the TME, chief among these cells are natural killer cells, neutrophils, and macrophages. Further research into tissue-based and non-invasive biomarkers that can be prospectively integrated into therapeutic trial design will be critical to advance the field's understanding of individual resistance patterns and enable the ultimate goal of precision immuno-oncology.
Collapse
Affiliation(s)
- Molly C Tokaz
- Division of Medical Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Christina S Baik
- Division of Medical Oncology, University of Washington, Seattle, WA, USA.,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - A McGarry Houghton
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.,Division of Pulmonology and Critical Care Medicine, University of Washington, Seattle, WA, USA.,Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Diane Tseng
- Division of Medical Oncology, University of Washington, Seattle, WA, USA. .,Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. .,Fred Hutchinson Cancer Center, Mail Stop LG-540, 825 Eastlake Ave E., Seattle, WA, 98109, USA.
| |
Collapse
|
3
|
Reticker-Flynn NE, Zhang W, Belk JA, Basto PA, Escalante NK, Pilarowski GOW, Bejnood A, Martins MM, Kenkel JA, Linde IL, Bagchi S, Yuan R, Chang S, Spitzer MH, Carmi Y, Cheng J, Tolentino LL, Choi O, Wu N, Kong CS, Gentles AJ, Sunwoo JB, Satpathy AT, Plevritis SK, Engleman EG. Lymph node colonization induces tumor-immune tolerance to promote distant metastasis. Cell 2022; 185:1924-1942.e23. [PMID: 35525247 PMCID: PMC9149144 DOI: 10.1016/j.cell.2022.04.019] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 01/31/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022]
Abstract
For many solid malignancies, lymph node (LN) involvement represents a harbinger of distant metastatic disease and, therefore, an important prognostic factor. Beyond its utility as a biomarker, whether and how LN metastasis plays an active role in shaping distant metastasis remains an open question. Here, we develop a syngeneic melanoma mouse model of LN metastasis to investigate how tumors spread to LNs and whether LN colonization influences metastasis to distant tissues. We show that an epigenetically instilled tumor-intrinsic interferon response program confers enhanced LN metastatic potential by enabling the evasion of NK cells and promoting LN colonization. LN metastases resist T cell-mediated cytotoxicity, induce antigen-specific regulatory T cells, and generate tumor-specific immune tolerance that subsequently facilitates distant tumor colonization. These effects extend to human cancers and other murine cancer models, implicating a conserved systemic mechanism by which malignancies spread to distant organs.
Collapse
Affiliation(s)
| | - Weiruo Zhang
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Julia A Belk
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Pamela A Basto
- Division of Oncology, Department of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | | | | | - Alborz Bejnood
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Maria M Martins
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Justin A Kenkel
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Ian L Linde
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Sreya Bagchi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Robert Yuan
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Serena Chang
- Institute for Immunity, Transplantation, and Infection Operations, Stanford University, Palo Alto, CA 94305, USA; Department of Otolaryngology-Head & Neck Surgery, Stanford University, Palo Alto, CA 94305, USA
| | - Matthew H Spitzer
- Department of Microbiology and Immunology and Department of Otolaryngology-Head and Neck Surgery, University of California, San Francisco, CA, USA
| | - Yaron Carmi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jiahan Cheng
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lorna L Tolentino
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Okmi Choi
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Nancy Wu
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Christina S Kong
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Palo Alto, CA 94305, USA
| | - John B Sunwoo
- Department of Otolaryngology-Head & Neck Surgery, Stanford University, Palo Alto, CA 94305, USA; Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA; Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Sylvia K Plevritis
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA; Department of Radiology, Stanford University, Palo Alto, CA 94305, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University, Palo Alto, CA 94305, USA.
| |
Collapse
|
4
|
Challenges of the Immunotherapy: Perspectives and Limitations of the Immune Checkpoint Inhibitor Treatment. Int J Mol Sci 2022; 23:ijms23052847. [PMID: 35269988 PMCID: PMC8910928 DOI: 10.3390/ijms23052847] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
Immunotherapy is a quickly developing type of treatment and the future of therapy in oncology. This paper is a review of recent findings in the field of immunotherapy with an emphasis on immune checkpoint inhibitors. The challenges that immunotherapy might face in near future, such as primary and acquired resistance and the irAEs, are described in this article, as well as the perspectives such as identification of environmental modifiers of immunity and development of anti-cancer vaccines and combined therapies. There are multiple factors that may be responsible for immunoresistance, such as genomic factors, factors related to the immune system cells or to the cancer microenvironment, factors emerging from the host cells, as well as other factors such as advanced age, biological sex, diet, many hormones, existing comorbidities, and the gut microbiome.
Collapse
|
5
|
Gao Y, Päivinen P, Tripathi S, Domènech-Moreno E, Wong IPL, Vaahtomeri K, Nagaraj AS, Talwelkar SS, Foretz M, Verschuren EW, Viollet B, Yan Y, Mäkelä TP. Inactivation of AMPK Leads to Attenuation of Antigen Presentation and Immune Evasion in Lung Adenocarcinoma. Clin Cancer Res 2021; 28:227-237. [PMID: 34667030 DOI: 10.1158/1078-0432.ccr-21-2049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/21/2021] [Accepted: 10/06/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Mutations in STK11 (LKB1) occur in 17% of lung adenocarcinoma (LUAD) and drive a suppressive (cold) tumor immune microenvironment (TIME) and resistance to immunotherapy. The mechanisms underpinning the establishment and maintenance of a cold TIME in LKB1-mutant LUAD remain poorly understood. In this study, we investigated the role of the LKB1 substrate AMPK in immune evasion in human non-small cell lung cancer (NSCLC) and mouse models and explored the mechanisms involved. EXPERIMENTAL DESIGN We addressed the role of AMPK in immune evasion in NSCLC by correlating AMPK phosphorylation and immune-suppressive signatures and by deleting AMPKα1 (Prkaa1) and AMPKα2 (Prkaa2) in a KrasG12D -driven LUAD. Furthermore, we dissected the molecular mechanisms involved in immune evasion by comparing gene-expression signatures, AMPK activity, and immune infiltration in mouse and human LUAD and gain or loss-of-function experiments with LKB1- or AMPK-deficient cell lines. RESULTS Inactivation of both AMPKα1 and AMPKα2 together with Kras activation accelerated tumorigenesis and led to tumors with reduced infiltration of CD8+/CD4+ T cells and gene signatures associated with a suppressive TIME. These signatures recapitulate those in Lkb1-deleted murine LUAD and in LKB1-deficient human NSCLC. Interestingly, a similar signature is noted in human NSCLC with low AMPK activity. In mechanistic studies, we find that compromised LKB1 and AMPK activity leads to attenuated antigen presentation in both LUAD mouse models and human NSCLC. CONCLUSIONS The results provide evidence that the immune evasion noted in LKB1-inactivated lung cancer is due to subsequent inactivation of AMPK and attenuation of antigen presentation.
Collapse
Affiliation(s)
- Yajing Gao
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Colorectal Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pekka Päivinen
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Sushil Tripathi
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Eva Domènech-Moreno
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Iris P L Wong
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Kari Vaahtomeri
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki and Wihuri Research Institute, Helsinki, Finland
| | - Ashwini S Nagaraj
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sarang S Talwelkar
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marc Foretz
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Emmy W Verschuren
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Benoit Viollet
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Yan Yan
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland. .,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland.,Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Tomi P Mäkelä
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland.,HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
6
|
Zhu S, Zhang T, Zheng L, Liu H, Song W, Liu D, Li Z, Pan CX. Combination strategies to maximize the benefits of cancer immunotherapy. J Hematol Oncol 2021; 14:156. [PMID: 34579759 PMCID: PMC8475356 DOI: 10.1186/s13045-021-01164-5] [Citation(s) in RCA: 308] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/07/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapies such as immune checkpoint blockade (ICB) and adoptive cell therapy (ACT) have revolutionized cancer treatment, especially in patients whose disease was otherwise considered incurable. However, primary and secondary resistance to single agent immunotherapy often results in treatment failure, and only a minority of patients experience long-term benefits. This review article will discuss the relationship between cancer immune response and mechanisms of resistance to immunotherapy. It will also provide a comprehensive review on the latest clinical status of combination therapies (e.g., immunotherapy with chemotherapy, radiation therapy and targeted therapy), and discuss combination therapies approved by the US Food and Drug Administration. It will provide an overview of therapies targeting cytokines and other soluble immunoregulatory factors, ACT, virotherapy, innate immune modifiers and cancer vaccines, as well as combination therapies that exploit alternative immune targets and other therapeutic modalities. Finally, this review will include the stimulating insights from the 2020 China Immuno-Oncology Workshop co-organized by the Chinese American Hematologist and Oncologist Network (CAHON), the China National Medical Product Administration (NMPA) and Tsinghua University School of Medicine.
Collapse
Affiliation(s)
- Shaoming Zhu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Department of Urology, Beijing Chao-Yang Hospital, Beijing, China
| | - Tian Zhang
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Division of Medical Oncology, Department of Medicine, Duke Cancer Institute, Duke University, DUMC 103861, Durham, NC, 27710, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,The Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,University of Chicago, Chicago, IL, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.,New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA.
| | - Chong-Xian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA. .,Harvard Medical School, West Roxbury, MA, 02132, USA.
| |
Collapse
|
7
|
Beta2-microglobulin(B2M) in cancer immunotherapies: Biological function, resistance and remedy. Cancer Lett 2021; 517:96-104. [PMID: 34129878 DOI: 10.1016/j.canlet.2021.06.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/30/2022]
Abstract
Cancer immunotherapies have made much headway during the past decades. Techniques including the immune checkpoint inhibition (ICI) and adoptive cell therapy (ACT) have harvested impressive efficacy and provided far-reaching tools for treating cancer patients. However, due to inadequate priming of the immune system, a certain subgroup of patients remains resistant to cancer immunotherapies during or after the treatment. β2-microglobulin (B2M) is an important subunit of major histocompatibility complex (MHC) class I which exerts substantive biological functions in tumorigenesis and immune control. Accumulating evidence has shown that alterations of B2M gene and B2M proteins contribute to poor reaction to cancer immunotherapies by dampening antigen presentation. Here, we discuss the basic biological functions of B2M, its distribution in a spectrum of cancers, and current understanding of its role in ICI, cancer vaccines and chimeric antigen receptor T cell (CAR-T) therapies. Furthermore, we summarize some promising therapeutic strategies to improve the efficacy inhibited by B2M defects.
Collapse
|
8
|
Zhao Y, Cao Y, Chen Y, Wu L, Hang H, Jiang C, Zhou X. B2M gene expression shapes the immune landscape of lung adenocarcinoma and determines the response to immunotherapy. Immunology 2021; 164:507-523. [PMID: 34115389 DOI: 10.1111/imm.13384] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/20/2021] [Accepted: 06/06/2021] [Indexed: 01/19/2023] Open
Abstract
Loss of the B2M gene is associated with tumour immune escape and resistance to immunotherapy. However, genetic alterations of the B2M gene are rare. We performed an integrative analysis of the mutational and transcriptional profiles of large cohorts of non-small-cell lung cancer (NSCLC) patients and found that epigenetic downregulation of B2M is common. B2M-low tumours exhibit a suppressive immune microenvironment characterized by reduced infiltration of immune cells of various lineages; in B2M-high tumours, more T and natural killer cells are present, but their activities are constrained by immune checkpoint molecules, indicating the diverse mechanisms of immune evasion. High levels of B2M mRNA, but not PD-L1, are correlated with an enhanced response to PD-1-based immunotherapy, suggesting its value for immunotherapy response prediction in solid tumours. Notably, a high tumour mutation burden (TMB) is associated with low B2M expression, which may explain the poor predictive value of the TMB in some situations. In syngeneic mouse models, genetic ablation of B2M in tumour cells causes resistance to PD-1-based immunotherapy, and B2M knockdown also diminishes the therapeutic efficacy. Moreover, forced expression of B2M in tumour models improves the response to immunotherapy, suggesting that B2M levels have significant impacts on treatment outcomes. Finally, we provide insight into the roles of transcription factors and KRAS mutations in B2M expression and the anticancer immune response. In conclusion, genetic and epigenetic regulation of B2M fundamentally shapes the NSCLC immune microenvironment and may determine the response to checkpoint blockade-based immunotherapy.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Yuejiao Cao
- School of Medicine, Nantong University, Nantong, China
| | - Yiqi Chen
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Lei Wu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Hua Hang
- Department of Pathology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Chenxia Jiang
- Department of Pathology, The Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.,Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, and Research Institution of Translational Medicine in Cardiothoracic Diseases in Affiliated Hospital of Nantong University, Jiangsu, China
| |
Collapse
|
9
|
Chi A, He X, Hou L, Nguyen NP, Zhu G, Cameron RB, Lee JM. Classification of Non-Small Cell Lung Cancer's Tumor Immune Micro-Environment and Strategies to Augment Its Response to Immune Checkpoint Blockade. Cancers (Basel) 2021; 13:cancers13122924. [PMID: 34208113 PMCID: PMC8230820 DOI: 10.3390/cancers13122924] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/03/2021] [Accepted: 06/09/2021] [Indexed: 12/26/2022] Open
Abstract
Simple Summary Immune checkpoint blockade (ICB) has become a major treatment for lung cancer. Better understanding of the tumor immune micro-environment (TIME) in non-small cell lung cancer (NSCLC) is urgently needed to better treat it with this type of therapy. In this review, we describe and explore how NSCLC’s TIME relates to response to ICB, as well as how to treat those with unresponsive types of TIME, which will significantly impact future research in lung cancer immunotherapy. Abstract Immune checkpoint blockade (ICB) with checkpoint inhibitors has led to significant and durable response in a subset of patients with advanced stage EGFR and ALK wild-type non-small cell lung cancer (NSCLC). This has been consistently shown to be correlated with the unique characteristics of each patient’s tumor immune micro-environment (TIME), including the composition and distribution of the tumor immune cell infiltrate; the expression of various checkpoints by tumor and immune cells, such as PD-L1; and the presence of various cytokines and chemokines. In this review, the classification of various types of TIME that are present in NSCLC and their correlation with response to ICB in NSCLC are discussed. This is conducted with a focus on the characteristics and identifiable biomarkers of different TIME subtypes that may also be used to predict NSCLC’s clinical response to ICB. Finally, treatment strategies to augment response to ICB in NSCLC with unresponsive types of TIME are explored.
Collapse
Affiliation(s)
- Alexander Chi
- Department of Radiation Oncology, Beijing Chest Hospital, Capital Medical University, Beijing 101100, China
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Xia He
- Department of Radiation Oncology, Jiangsu Cancer Hospital, Nanjing Medical University, Nanjing 210009, China
- Correspondence: (A.C.); (X.H.)
| | - Lin Hou
- Center for Statistical Science, Tsinghua University, Beijing 100084, China;
| | - Nam P. Nguyen
- Department of Radiation Oncology, Howard University, Washington, DC 20060, USA;
| | - Guangying Zhu
- Department of Radiation Oncology, China-Japan Friendship Hospital, Beijing 100029, China;
| | - Robert B. Cameron
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| | - Jay M. Lee
- Division of Thoracic Surgery, Department of Surgery, University of California at Los Angeles, Los Angeles, CA 90095, USA; (R.B.C.); (J.M.L.)
| |
Collapse
|
10
|
Tabassum A, Samdani MN, Dhali TC, Alam R, Ahammad F, Samad A, Karpiński TM. Transporter associated with antigen processing 1 (TAP1) expression and prognostic analysis in breast, lung, liver, and ovarian cancer. J Mol Med (Berl) 2021; 99:1293-1309. [PMID: 34047812 PMCID: PMC8367907 DOI: 10.1007/s00109-021-02088-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 04/15/2021] [Accepted: 05/05/2021] [Indexed: 12/25/2022]
Abstract
Abstract Transporter associated with antigen processing 1 (TAP1) is a transporter protein that represent tumor antigen in the MHC I or HLA complex. Any defect in the TAP1 gene resulting in inadequate tumor tracking. TAP1 influences multidrug resistance (MDR) in human cancer cell lines and hinders the treatment during chemotherapeutic. The association of TAP1 in cancer progression remains mostly unknown and further study of the gene in relation with cancer need to conduct. Thus, the study has designed to analyze the association between the TAP1 with cancer by computationally. The expression pattern of the gene has determined by using ONCOMINE, GENT2, and GEPIA2 online platforms. The protein level of TAP1 was examined by the help of Human Protein Atlas. Samples with different clinical outcomes were investigated to evaluate the expression and promoter methylation in cancer vs. normal tissues by using UALCAN server. The copy number alteration, mutation frequency, and expression level of the gene in different cancer were analyzed by using cBioPortal server. The PrognoScan and KM plotter platforms were used to perform the survival analysis and represented graphically. Additionally, pathway and gene ontology (GO) features correlated to the TAP1 gene were analyzed and presented by bar charts. After arranging the data in a single panel like correlating expression to prognosis, mutational and alterations characteristic, and pathways analysis, we observed some interesting insights that emphasized the importance of the gene in cancer progression. The study found the relationship between the TAP1 expression pattern and prognosis in different cancer tissues and shows how TAP1 affects the clinical characteristics. The analytical data presented in the study is vital to learn about the effect of TAP1 in tumor tissue, where previously studies showing contradicting expression of TAP1 in cancer tissue. The analyzed data can also be utilized further to evade the threats against chemotherapy. Overall, the study provided a new aspect to consider the role of TAP1 gene in cancer progression and survival status. Key messages • This study demonstrated, for the first time, a correlation between the TAP1 gene and tumor progression. • An upregulation of TAP1 mRNA was demonstrated in various cancer types. • This study reported a significant negative correlation for TAP1 gene expression and the survival rate in different cancer types. Supplementary Information The online version contains supplementary material available at 10.1007/s00109-021-02088-w.
Collapse
Affiliation(s)
- Anika Tabassum
- Biochemistry Department, School of Life Sciences, Independent University, Dhaka, 1229, Bangladesh
| | - Md Nazmus Samdani
- Department of Pharmacy, University of Dhaka, Dhaka, 1000, Bangladesh
| | - Tarak Chandra Dhali
- Department of Biotechnology and Genetic Engineering, Khulna University, Khulna, 9208, Bangladesh
| | - Rahat Alam
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh.,Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Foysal Ahammad
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh. .,Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh. .,Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, 21589, Saudi Arabia.
| | - Abdus Samad
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh. .,Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh.
| | - Tomasz M Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Wieniawskiego 3, 61-712, Poznań, Poland.
| |
Collapse
|
11
|
Politi K. Leveraging Patient-Derived Models for Immunotherapy Research. Am Soc Clin Oncol Educ Book 2021; 40:e344-e350. [PMID: 32511030 DOI: 10.1200/edbk_280579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
As cancer immunotherapies become mainstream for the treatment of many different cancer types and the numbers of new agents continue to increase, the need for experimental models is also rising. An approach to develop and study models for immune-oncology that has garnered intense interest in recent years is that of using patient-derived models. Patient-derived models can recapitulate many of the features and heterogeneity of the corresponding human tumors. Historically these models have been used to study cancer cell-intrinsic properties of tumors and drugs that target tumor cells directly. In recent years, increasing recognition of the role immune cells play in cancer and how these represent good therapeutic targets has led to efforts to optimize and use patient-derived models for cancer immunotherapy studies. Patient-derived models are now being used to study the properties of cancer cells that modulate their ability to respond to immune stimulation. Further efforts are underway to use and develop patient-derived models that incorporate human immune cells in vitro and in vivo (humanized mice) so that cancer cell-immune cell interactions can be studied in the context of cancer immunotherapies. As these models are further refined, leveraging patient-derived models for cancer immunotherapy research can provide insight into mechanisms of sensitivity and resistance to cancer immunotherapies, uncover new targets, reveal how specific agents work, and be used to evaluate the antitumor efficacy of therapeutic regimens.
Collapse
Affiliation(s)
- Katerina Politi
- Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
12
|
Weng CY, Kao CX, Chang TS, Huang YH. Immuno-Metabolism: The Role of Cancer Niche in Immune Checkpoint Inhibitor Resistance. Int J Mol Sci 2021; 22:1258. [PMID: 33514004 PMCID: PMC7865434 DOI: 10.3390/ijms22031258] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICI) in treating cancer has revolutionized the approach to eradicate cancer cells by reactivating immune responses. However, only a subset of patients benefits from this treatment; the majority remains unresponsive or develops resistance to ICI therapy. Increasing evidence suggests that metabolic machinery in the tumor microenvironment (TME) plays a role in the development of ICI resistance. Within the TME, nutrients and oxygen are scarce, forcing immune cells to undergo metabolic reprogramming to adapt to harsh conditions. Cancer-induced metabolic deregulation in immune cells can attenuate their anti-cancer properties, but can also increase their immunosuppressive properties. Therefore, targeting metabolic pathways of immune cells in the TME may strengthen the efficacy of ICIs and prevent ICI resistance. In this review, we discuss the interactions of immune cells and metabolic alterations in the TME. We also discuss current therapies targeting cellular metabolism in combination with ICIs for the treatment of cancer, and provide possible mechanisms behind the cellular metabolic rewiring that may improve clinical outcomes.
Collapse
Affiliation(s)
- Chao-Yuan Weng
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Cheng-Xiang Kao
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
13
|
Shi Y, Su H, Song Y, Jiang W, Sun X, Qian W, Zhang W, Gao Y, Jin Z, Zhou J, Jin C, Zou L, Qiu L, Li W, Yang J, Hou M, Xiong Y, Zhou H, Du X, Wang X, Peng B. Circulating tumor DNA predicts response in Chinese patients with relapsed or refractory classical hodgkin lymphoma treated with sintilimab. EBioMedicine 2021; 54:102731. [PMID: 32304999 PMCID: PMC7186760 DOI: 10.1016/j.ebiom.2020.102731] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 02/17/2020] [Accepted: 03/05/2020] [Indexed: 02/08/2023] Open
Abstract
Background Blood-based biomarker such as circulating tumor DNA (ctDNA) has emerged as a promising tool for assessment of response to immunotherapy in solid tumors; But in hematological malignances, evidences are still lacking to support its clinical utility. In current study the feasibility of ctDNA for prediction and monitoring of response to anti-PD-1 therapy in Chinese patients with relapsed or refractory classical Hodgkin lymphoma (r/r cHL) was assessed. Methods A total of 192 plasma samples from 75 patients with r/r cHL were collected at baseline and upon therapeutic evaluation. ctDNA were sequenced by targeting panels capturing frequently mutated genes in cHL and other hematological malignancies and then quantified. Analysis on: 1) Gene mutation profile and association of the gene mutations with progression-free survival; 2) Association of pre- and post-treatment ctDNA variant allelic frequencies with clinical outcome; (3) Correlation of the mutated genes with treatment resistance; were performed. Findings Somatic mutations were detected in 50 out of 61 patients by ctDNA genotyping. The mutations of CHD8 was significantly higher in patients with PFS ≥ 12 months. Baseline ctDNA was significantly higher in responders and a decrease of ctDNA ≥ 40% from baseline indicated superior clinical outcome. Strong agreement between ctDNA dynamic and radiographic response change during therapy was observed in majority of the patients. Furthermore, the mutations of B2M, TNFRSF14 and KDM2B were found to be associated with acquired resistance. Interpretation ctDNA could be an informative biomarker for anti-PD-1 immunotherapy in r/r cHL. Funding This work was supported by Innovent Biologics, Eli Lilly and Companyhttps://doi.org/10.13039/501100002852, China National New Drug Innovation Program (2014ZX09201041-001 and 2017ZX09304015), Chinese Academy of Medical Sciences (CAMS) Innovation Fund for Medical Sciences (CIFMS) (2016-I2M-1-001) and National Key Scientific Program Precision Medicine Research Fund of China (2017YFC0909801). The funders had no role in study design, data collection, data analysis, interpretation or writing.
Collapse
Affiliation(s)
- Yuankai Shi
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| | - Hang Su
- The 307th Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yongping Song
- The affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Wenqi Jiang
- Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiuhua Sun
- Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wenbin Qian
- The First Affiliated Hospital, Medical School of Zhejiang University, Hangzhou, China
| | - Wei Zhang
- Peking Union Medical College Hospital, Beijing, China
| | - Yuhuan Gao
- Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhengming Jin
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianfeng Zhou
- Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuan Jin
- Cancer Hospital Affiliated to Guangzhou Medical University, Guangzhou, China
| | - Liqun Zou
- West China Hospital, Sichuan University, Chengdu, China
| | - Lugui Qiu
- Blood Institute of Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Wei Li
- The First Hospital of Jilin University, Changchun, China
| | | | - Ming Hou
- Qilu Hospital of Shandong University, Jinan, China
| | - Yan Xiong
- Innovent Biologics (Suzhou) Co., Ltd, China
| | - Hui Zhou
- Innovent Biologics (Suzhou) Co., Ltd, China
| | | | - Xiong Wang
- Innovent Biologics (Suzhou) Co., Ltd, China
| | - Bo Peng
- Innovent Biologics (Suzhou) Co., Ltd, China
| |
Collapse
|
14
|
Hu-Lieskovan S, Malouf GG, Jacobs I, Chou J, Liu L, Johnson ML. Addressing resistance to immune checkpoint inhibitor therapy: an urgent unmet need. Future Oncol 2021; 17:1401-1439. [PMID: 33475012 DOI: 10.2217/fon-2020-0967] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of various cancers by reversing the immunosuppressive mechanisms employed by tumors to restore anticancer immunity. Although ICIs have demonstrated substantial clinical efficacy, patient response can vary in depth and duration, and many do not respond at all or eventually develop resistance. ICI resistance mechanisms can be tumor-intrinsic, related to the tumor microenvironment or patient-specific factors. Multiple resistance mechanisms may be present within one tumor subtype, or heterogeneity exists among patients with the same tumor type. Consequently, designing effective combination treatment strategies is challenging. This review will discuss ICI resistance mechanisms, and summarize findings from key preclinical and clinical trials of ICIs, to identify potential treatment strategies or pathways to overcome ICI resistance.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Department of Medicine, Division of Oncology, Huntsman Cancer Institute / University of Utah, Salt Lake City, UT 84112, USA
| | - Gabriel G Malouf
- Department of Medical Oncology, Institut de Cancérologie de Strasbourg & Department of Functional Genomics & Cancer, Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS/INSERM/UNISTRA, Illkirch Cedex, Strasbourg, France
| | | | | | - Li Liu
- Pfizer Inc, San Diego, CA 92121, USA
| | - Melissa L Johnson
- Sarah Cannon Research Institute/Tennessee Oncology, PLLC, Nashville, TN 37203, USA
| |
Collapse
|
15
|
Pros E, Saigi M, Alameda D, Gomez-Mariano G, Martinez-Delgado B, Alburquerque-Bejar JJ, Carretero J, Tonda R, Esteve-Codina A, Catala I, Palmero R, Jove M, Lazaro C, Patiño-Garcia A, Gil-Bazo I, Verdura S, Teulé A, Torres-Lanzas J, Sidransky D, Reguart N, Pio R, Juan-Vidal O, Nadal E, Felip E, Montuenga LM, Sanchez-Cespedes M. Genome-wide profiling of non-smoking-related lung cancer cells reveals common RB1 rearrangements associated with histopathologic transformation in EGFR-mutant tumors. Ann Oncol 2021; 31:274-282. [PMID: 31959344 DOI: 10.1016/j.annonc.2019.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND The etiology and the molecular basis of lung adenocarcinomas (LuADs) in nonsmokers are currently unknown. Furthermore, the scarcity of available primary cultures continues to hamper our biological understanding of non-smoking-related lung adenocarcinomas (NSK-LuADs). PATIENTS AND METHODS We established patient-derived cancer cell (PDC) cultures from metastatic NSK-LuADs, including two pairs of matched EGFR-mutant PDCs before and after resistance to tyrosine kinase inhibitors (TKIs), and then performed whole-exome and RNA sequencing to delineate their genomic architecture. For validation, we analyzed independent cohorts of primary LuADs. RESULTS In addition to known non-smoker-associated alterations (e.g. RET, ALK, EGFR, and ERBB2), we discovered novel fusions and recurrently mutated genes, including ATF7IP, a regulator of gene expression, that was inactivated in 5% of primary LuAD cases. We also found germline mutations at dominant familiar-cancer genes, highlighting the importance of genetic predisposition in the origin of a subset of NSK-LuADs. Furthermore, there was an over-representation of inactivating alterations at RB1, mostly through complex intragenic rearrangements, in treatment-naive EGFR-mutant LuADs. Three EGFR-mutant and one EGFR-wild-type tumors acquired resistance to EGFR-TKIs and chemotherapy, respectively, and histology on re-biopsies revealed the development of small-cell lung cancer/squamous cell carcinoma (SCLC/LuSCC) transformation. These features were consistent with RB1 inactivation and acquired EGFR-T790M mutation or FGFR3-TACC3 fusion in EGFR-mutant tumors. CONCLUSIONS We found recurrent alterations in LuADs that deserve further exploration. Our work also demonstrates that a subset of NSK-LuADs arises within cancer-predisposition syndromes. The preferential occurrence of RB1 inactivation, via complex rearrangements, found in EGFR-mutant tumors appears to favor SCLC/LuSCC transformation under growth-inhibition pressures. Thus RB1 inactivation may predict the risk of LuAD transformation to a more aggressive type of lung cancer, and may need to be considered as a part of the clinical management of NSK-LuADs patients.
Collapse
Affiliation(s)
- E Pros
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - M Saigi
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain; Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - D Alameda
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra and Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - G Gomez-Mariano
- Molecular Genetics Unit, Rare Diseases Institute of Research (IIER), Health Institute Carlos III, Majadahonda, Madrid, Spain
| | - B Martinez-Delgado
- Molecular Genetics Unit, Rare Diseases Institute of Research (IIER), Health Institute Carlos III, Majadahonda, Madrid, Spain
| | - J J Alburquerque-Bejar
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - J Carretero
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, Valencia, Spain
| | - R Tonda
- CNAG-CRG, Centre for Genomic Regulation (CRG), Institute of Science and Technology (BIST) and University Pompeu Fabra (UPF), Barcelona, Spain
| | - A Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Institute of Science and Technology (BIST) and University Pompeu Fabra (UPF), Barcelona, Spain
| | - I Catala
- Pathology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Spain
| | - R Palmero
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - M Jove
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain
| | - C Lazaro
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO-Oncobell-IDIBELL, Madrid, Spain; CIBERONC, Health Institute Carlos III, Madrid, Spain
| | - A Patiño-Garcia
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra and Navarra Health Research Institute (IDISNA), Pamplona, Spain
| | - I Gil-Bazo
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra and Navarra Health Research Institute (IDISNA), Pamplona, Spain; CIBERONC, Health Institute Carlos III, Madrid, Spain
| | - S Verdura
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - A Teulé
- Hereditary Cancer Program, Catalan Institute of Oncology, ICO-Oncobell-IDIBELL, Madrid, Spain; CIBERONC, Health Institute Carlos III, Madrid, Spain
| | - J Torres-Lanzas
- Thoracic Surgery Department, Hospital Universitario Virgen de la Arrixaca, El Palmar-Murcia, Spain
| | - D Sidransky
- Department of Otolaryngology and Head and Neck Surgery, Johns Hopkins University, School of Medicine, Baltimore, Maryland
| | - N Reguart
- Oncology Department, Thoracic Tumors Unit, Clinic Hospital, Barcelona, Spain; Translational Genomics and Targeted Therapeutics in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - R Pio
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra and Navarra Health Research Institute (IDISNA), Pamplona, Spain; CIBERONC, Health Institute Carlos III, Madrid, Spain
| | - O Juan-Vidal
- Biomarkers and Precision Medicine Unit, Research Institute La Fe and Department of Medical Oncology, La Fe University Hospital, Valencia, Spain
| | - E Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain; Clinical Research in Solid Tumors Group (CReST), Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - E Felip
- Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - L M Montuenga
- Program in Solid Tumors, Center for Applied Medical Research (CIMA), University of Navarra and Navarra Health Research Institute (IDISNA), Pamplona, Spain; CIBERONC, Health Institute Carlos III, Madrid, Spain; Department of Pathology, Anatomy and Physiology, Schools of Medicine and Sciences, University of Navarra, Pamplona, Spain
| | - M Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
16
|
Lagos GG, Izar B, Rizvi NA. Beyond Tumor PD-L1: Emerging Genomic Biomarkers for Checkpoint Inhibitor Immunotherapy. Am Soc Clin Oncol Educ Book 2020; 40:1-11. [PMID: 32315237 DOI: 10.1200/edbk_289967] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Despite the success of immune checkpoint blockade as a strategy for activating an antitumor immune response and promoting cancer regression, only a subset of patients have durable clinical benefit. Efforts are ongoing to identify robust biomarkers that can effectively predict treatment response to immune checkpoint inhibitors (ICIs). Although PD-L1 expression is useful for stratifying patients, it is an imperfect tool. Comprehensive next-generation sequencing platforms that are readily used in clinical practice to identify a tumor's potentially actionable genetic alterations also reveal tumor genomic features, including tumor mutation burden (TMB), that may impact the response to ICIs. High TMB enhances tumor immunogenicity through increased numbers of tumor neoantigens that may promote an immune response. Defective DNA repair, leading to microsatellite instability, is an endogenous mechanism for increased tumor TMB that augments response to anti-PD-1 blockade. Alternatively, DNA damage from exogenous factors is responsible for high TMB seen in melanoma, lung cancer, and urothelial carcinoma, among tumor subtypes with higher response rates to ICIs. In this review, we summarize data supporting the use of TMB as a biomarker as well as its known limitations. We also highlight specific tumor suppressor genes and oncogenes that are under investigation as biomarkers for ICI response and resistance. Efforts are ongoing to delineate which genomic tumor characteristics can eventually be utilized in clinical practice to ascertain the benefit of ICIs for an individual patient.
Collapse
|
17
|
Abstract
Immune checkpoint inhibitors (ICIs), monoclonal antibodies to cytotoxic T-lymphocyte-associated protein 4, programmed cell death 1 or its ligand PD-L1 are rapidly changing the treatment landscape and prognosis of many cancer types. Following their initial approval in melanoma in 2011, ICIs are now approved in many other cancers. Despite the long-term, durable response that can be noted with ICIs, the majority of patients do not respond to ICIs and some of the initial responders develop relapsed disease during their treatment course. In order to improve the response rate to ICIs, an understanding of the mechanisms of resistance is critical. Given the number of different ways cancers can become resistant to ICIs, patient-rather than population-based strategies to reverse resistance will likely be needed. We review the currently defined mechanisms of resistance to ICIs and discuss possible methods to overcome these mechanisms.
Collapse
Affiliation(s)
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
18
|
Calmeiro J, Carrascal MA, Tavares AR, Ferreira DA, Gomes C, Cruz MT, Falcão A, Neves BM. Pharmacological combination of nivolumab with dendritic cell vaccines in cancer immunotherapy: An overview. Pharmacol Res 2020; 164:105309. [PMID: 33212291 DOI: 10.1016/j.phrs.2020.105309] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/24/2020] [Accepted: 10/25/2020] [Indexed: 02/06/2023]
Abstract
In the last decade, immunotherapy led to a paradigm shift in the treatment of numerous malignancies. Alongside with monoclonal antibodies blocking programmed cell death receptor-1 (PD-1)/PD-L1 and cytotoxic T- lymphocyte antigen 4 (CTLA-4) immune checkpoints, cell-based approaches such as CAR-T cells and dendritic cell (DC) vaccines have strongly contributed to pushing forward this thrilling field. While initial strategies were mainly focused on monotherapeutic regimens, it is now consensual that the combination of immunotherapies tackling multiple cancer hallmarks can result in superior clinical outcomes. Here, we review in depth the pharmacological combination of DC-based vaccines that boost tumour elimination by eliciting and expanding effector immune cells, with the PD-1 inhibitor Nivolumab that allows blocking key tumour immune escape mechanisms. This combination represents an important step in cancer therapy, with a significant enhancement in patient survival in several types of tumours, paving an important way in establishing combinatorial immunotherapeutic strategies as first-line treatments.
Collapse
Affiliation(s)
- João Calmeiro
- Faculty of Pharmacy, FFUC, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mylène A Carrascal
- Center for Neuroscience and Cell Biology, CNC, University of Coimbra, 3004-504, Coimbra, Portugal; Tecnimede Group, 2710-089, Sintra, Portugal
| | - Adriana Ramos Tavares
- Faculty of Pharmacy, FFUC, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Daniel Alexandre Ferreira
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, 300-504, Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, FFUC, University of Coimbra, 3000-548, Coimbra, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, 3000-548, Coimbra, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, 3000-548, Coimbra, Portugal.
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
19
|
Zhang X, Liu M, Zhang X, Wang Y, Dai L. Autoantibodies to tumor-associated antigens in lung cancer diagnosis. Adv Clin Chem 2020; 103:1-45. [PMID: 34229848 DOI: 10.1016/bs.acc.2020.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lung cancer (LC) accounts for the majority of cancer-related deaths worldwide. Although screening the high-risk population by low-dose CT (LDCT) has reduced mortality, the cost and high false positivity rate has prevented its general diagnostic use. As such, better and more specific minimally invasive biomarkers are needed in general and for early LC detection, specifically. Autoantibodies produced by humoral immune response to tumor-associated antigens (TAA) are emerging as a promising noninvasive biomarker for LC. Given the low sensitivity of any one single autoantibody, a panel approach could provide a more robust and promising strategy to detect early stage LC. In this review, we summarize the background of TAA autoantibodies (TAAb) and the techniques currently used for identifying TAA, as well as recent findings of LC specific antigens and TAAb. This review provides guidance toward the development of accurate and reliable TAAb as immunodiagnostic biomarkers in the early detection of LC.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan, China
| | - Man Liu
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Xue Zhang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Yulin Wang
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences & Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Tumor Epidemiology, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Sun Y, Yu H, Li F, Lan L, He D, Zhao H, Qi D. Identification of Hub Genes and Potential Molecular Mechanisms in Patients with HBV-Associated Acute Liver Failure. Evol Bioinform Online 2020; 16:1176934320943901. [PMID: 33100826 PMCID: PMC7549162 DOI: 10.1177/1176934320943901] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Hepatitis B virus (HBV) infection is a major cause of acute liver failure (ALF) in China, and mortality rates are high among patients who do not receive a matched liver transplant. This study aimed to determine potential mechanisms involved in HBV-ALF pathogenesis. Gene expression profiles under access numbers GSE38941 and GSE14668 were downloaded from the Gene Expression Omnibus database, including cohorts of HBV-ALF liver tissue and normal samples. Differentially expressed genes (DEGs) with false discovery rates (FDR) <0.05 and |log2(fold change)| >1 as thresholds were screened using the Limma package. Gene modules associated with stable disease were mined using weighed gene co-expression network analysis (WGCNA). A co-expression network was constructed and DEGs were analyzed using gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. A gene-based network was constructed to explore major factors associated with disease progression. We identified 2238 overlapping DEGs as crucial gene cohorts in ALF development. Based on a WGCNA algorithm, 10 modules (modules 1-10) were obtained that ranged from 75 to 1078 genes per module. Cyclin-dependent kinase 1 (CDK1), cyclin B1 (CCNB1), and cell-division cycle protein 20 (CDC20) hub genes were screened using the co-expression network. Furthermore, 17 GO terms and 6 KEGG pathways were identified, such as cell division, immune response process, and antigen processing and presentation. Two overlapping signaling pathways that are crucial factors in HBV-ALF were screened using the Comprehensive Toxicogenomics Database (CTD). Several candidate genes including HLA-E, B2M, HLA-DPA1, and SYK were associated with HBV-ALF progression. Natural killer cell-mediated cytotoxicity and antigen presentation contributed to the progression of HBV-ALF. The HLA-E, B2M, HLA-DPA1, and SYK genes play critical roles in the pathogenesis and development of HBV-ALF.
Collapse
Affiliation(s)
- Ying Sun
- Department of Gastroenterology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Haitao Yu
- Intensive Care Unit, Qingdao Municipal Hospital, Qingdao, China
| | - Fangfang Li
- Department of Respiratory Medicine, Qingdao Eighth People's Hospital, Qingdao, China
| | - Liqiang Lan
- Department of Endocrinology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Daxin He
- Department of Ultrasound, Qingdao Municipal Hospital, Qingdao, China
| | - Haijun Zhao
- Department of Urology, Qingdao Municipal Hospital, Qingdao, China
| | - Dachuan Qi
- Department of Surgery, Shanghai Fourth People's Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
21
|
Thompson JC, Davis C, Deshpande C, Hwang WT, Jeffries S, Huang A, Mitchell TC, Langer CJ, Albelda SM. Gene signature of antigen processing and presentation machinery predicts response to checkpoint blockade in non-small cell lung cancer (NSCLC) and melanoma. J Immunother Cancer 2020; 8:jitc-2020-000974. [PMID: 33028693 PMCID: PMC7542663 DOI: 10.1136/jitc-2020-000974] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/18/2020] [Indexed: 12/31/2022] Open
Abstract
Background Limited data exist on the role of alterations in HLA Class I antigen processing and presentation machinery in mediating response to immune checkpoint blockade (ICB). Methods This retrospective cohort study analyzed transcriptional profiles from pre-treatment tumor samples of 51 chemotherapy-refractory advanced non-small cell lung cancer (NSCLC) patients and two independent melanoma cohorts treated with ICB. An antigen processing machinery (APM) score was generated utilizing eight genes associated with APM (B2M, CALR, NLRC5, PSMB9, PSME1, PSME3, RFX5, and HSP90AB1). Associations were made for therapeutic response, progression-free survival (PFS) and overall survival (OS). Results In NSCLC, the APM score was significantly higher in responders compared with non-responders (p=0.0001). An APM score above the median value for the cohort was associated with improved PFS (HR 0.34 (0.18 to 0.64), p=0.001) and OS (HR 0.44 (0.23 to 0.83), p=0.006). The APM score was correlated with an inflammation score based on the established T-cell-inflamed resistance gene expression profile (Pearson’s r=0.58, p<0.0001). However, the APM score better predicted response to ICB relative to the inflammation score with area under a receiving operating characteristics curve of 0.84 and 0.70 for PFS and OS, respectively. In a cohort of 14 high-risk resectable stage III/IV melanoma patients treated with neoadjuvant anti-PD1 ICB, a higher APM score was associated with improved disease-free survival (HR: 0.08 (0.01 to 0.50), p=0.0065). In an additional independent melanoma cohort of 27 metastatic patients treated with ICB, a higher APM score was associated with improved OS (HR 0.29 (0.09 to 0.89), p=0.044). Conclusion Our data demonstrate that defects in antigen presentation may be an important feature in predicting outcomes to ICB in both lung cancer and melanoma.
Collapse
Affiliation(s)
- Jeffrey C Thompson
- Pulmonary and Critical Care, Thoracic Oncology Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Christiana Davis
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Charuhas Deshpande
- Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Wei-Ting Hwang
- Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seth Jeffries
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Alexander Huang
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Tara C Mitchell
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Corey J Langer
- Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Steven M Albelda
- Pulmonary and Critical Care, Thoracic Oncology Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
Horvath L, Thienpont B, Zhao L, Wolf D, Pircher A. Overcoming immunotherapy resistance in non-small cell lung cancer (NSCLC) - novel approaches and future outlook. Mol Cancer 2020; 19:141. [PMID: 32917214 PMCID: PMC7488475 DOI: 10.1186/s12943-020-01260-z] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy (IO) has revolutionized the therapy landscape of non-small cell lung cancer (NSCLC), significantly prolonging the overall survival (OS) of advanced stage patients. Over the recent years IO therapy has been broadly integrated into the first-line setting of non-oncogene driven NSCLC, either in combination with chemotherapy, or in selected patients with PD-L1high expression as monotherapy. Still, a significant proportion of patients suffer from disease progression. A better understanding of resistance mechanisms depicts a central goal to avoid or overcome IO resistance and to improve patient outcome.We here review major cellular and molecular pathways within the tumor microenvironment (TME) that may impact the evolution of IO resistance. We summarize upcoming treatment options after IO resistance including novel IO targets (e.g. RIG-I, STING) as well as interesting combinational approaches such as IO combined with anti-angiogenic agents or metabolic targets (e.g. IDO-1, adenosine signaling, arginase). By discussing the fundamental mode of action of IO within the TME, we aim to understand and manage IO resistance and to seed new ideas for effective therapeutic IO concepts.
Collapse
MESH Headings
- Arginase/genetics
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/therapy
- DEAD Box Protein 58/antagonists & inhibitors
- DEAD Box Protein 58/genetics
- DEAD Box Protein 58/immunology
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Humans
- Immunotherapy/adverse effects
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Receptors, Immunologic/antagonists & inhibitors
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Lena Horvath
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Liyun Zhao
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
- Medical Clinic III, Department of Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn (UKB), Sigmund-Freud-Street 25, 53127, Bonn, Germany
| | - Andreas Pircher
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
| |
Collapse
|
23
|
Biomarkers for immune checkpoint therapy targeting programmed death 1 and programmed death ligand 1. Biomed Pharmacother 2020; 130:110621. [PMID: 34321165 DOI: 10.1016/j.biopha.2020.110621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Rapidly increasing usages of immune checkpoint therapy for cancer treatment, particularly monoclonal antibodies that target programmed cell death-1 (PD-1) and its ligand PD-L1, have been achieved due to startling durable therapeutic efficacy with limited toxicity. The therapeutics significantly prolonged the overall survival and progression free survival of patients across multiple cancer types. However, the objective response rate of patients receiving this kind of treatment is substantially low. Therefore, it is of great importance to exploit reliable biomarkers that can robustly predict the therapeutic effects. Several biomarkers have been characterized for the selection of patients, which is mainly based on immunological and genetic criteria. Herein, we focus on the current progress regarding the biomarkers for anti-PD-1/PD-L1 therapy.
Collapse
|
24
|
Bai R, Chen N, Li L, Du N, Bai L, Lv Z, Tian H, Cui J. Mechanisms of Cancer Resistance to Immunotherapy. Front Oncol 2020; 10:1290. [PMID: 32850400 PMCID: PMC7425302 DOI: 10.3389/fonc.2020.01290] [Citation(s) in RCA: 180] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Over the last decade, based on the extensive development of preclinical animal studies and clinical trials, the efficacy, and mechanisms of immunotherapy have been fully explored. Significant and lasting clinical responses with immunotherapy provide a new breakthrough treatment for a variety of refractory cancer histologies, which gradually change the treatment pattern of tumors. However, although immune checkpoint inhibitor drugs are promising for achieving longer-term efficacy, their benefits in the overall population are still very low, such as low frequency of response in some common tumor types such as breast and prostate, and heterogeneity in the degree of response among different tumor lesions in the same patient, making immunotherapy with many limitations and challenges. Most patients do not respond to immunotherapy or inevitably develop resistance to treatment after a period of treatment, manifesting with primary resistance or acquired resistance who initially respond to treatment. The mechanisms of tumor immune resistance are very complex and involve multiple aspects such as genes, metabolism, inflammation, and abnormal neovascularization. Currently, many mechanisms of immunotherapy resistance have been characterized, and more continue to be uncovered. These efforts can improve the quality of medical care for cancer diagnosis and treatment, which improve the quality of life of patients, and finally lead to accurate individualized treatment. This review discusses mechanisms of cancer immunotherapy resistance including tumor-intrinsic factors and tumor-extrinsic factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
25
|
Rijensky NM, Blondheim Shraga NR, Barnea E, Peled N, Rosenbaum E, Popovtzer A, Stemmer SM, Livoff A, Shlapobersky M, Moskovits N, Perry D, Rubin E, Haviv I, Admon A. Identification of Tumor Antigens in the HLA Peptidome of Patient-derived Xenograft Tumors in Mouse. Mol Cell Proteomics 2020; 19:1360-1374. [PMID: 32451349 PMCID: PMC8015002 DOI: 10.1074/mcp.ra119.001876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
Personalized cancer immunotherapy targeting patient-specific cancer/testis antigens (CTA) and neoantigens may benefit from large-scale tumor human leukocyte antigen (HLA) peptidome (immunopeptidome) analysis, which aims to accurately identify antigens presented by tumor cells. Although significant efforts have been invested in analyzing the HLA peptidomes of fresh tumors, it is often impossible to obtain sufficient volumes of tumor tissues for comprehensive HLA peptidome characterization. This work attempted to overcome some of these obstacles by using patient-derived xenograft tumors (PDX) in mice as the tissue sources for HLA peptidome analysis. PDX tumors provide a proxy for the expansion of the patient tumor by re-grafting them through several passages to immune-compromised mice. The HLA peptidomes of human biopsies were compared with those derived from PDX tumors. Larger HLA peptidomes were obtained from the significantly larger PDX tumors as compared with the patient biopsies. The HLA peptidomes of different PDX tumors derived from the same source tumor biopsy were very reproducible, even following subsequent passages to new naïve mice. Many CTA-derived HLA peptides were discovered, as well as several potential neoantigens/variant sequences. Taken together, the use of PDX tumors for HLA peptidome analysis serves as a highly expandable and stable source of reproducible and authentic peptidomes, opening up new opportunities for defining large HLA peptidomes when only small tumor biopsies are available. This approach provides a large source for tumor antigens identification, potentially useful for personalized immunotherapy.
Collapse
Affiliation(s)
| | | | - Eilon Barnea
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel
| | - Nir Peled
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Eli Rosenbaum
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Aron Popovtzer
- Institute of Oncology, Davidoff Center, Rabin Medical Center and Sackler Faculty of Medicine, Tel-Aviv University, Petah Tikva, Israel
| | - Solomon M Stemmer
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alejandro Livoff
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Mark Shlapobersky
- Institute of Pathology, Barzilai University Medical Center, Ashkelon, Israel
| | - Neta Moskovits
- Davidoff Center, Rabin Medical Center, Beilinson Campus, Petach Tikva, and Felsentien medical research center, Petach Tikva, Israel
| | - Dafna Perry
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Eitan Rubin
- Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheva, Israel; The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Itzhak Haviv
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Arie Admon
- Department of Biology, Technion-Israel Institute of Technology Haifa, Israel.
| |
Collapse
|
26
|
Lei Q, Wang D, Sun K, Wang L, Zhang Y. Resistance Mechanisms of Anti-PD1/PDL1 Therapy in Solid Tumors. Front Cell Dev Biol 2020; 8:672. [PMID: 32793604 PMCID: PMC7385189 DOI: 10.3389/fcell.2020.00672] [Citation(s) in RCA: 230] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 07/02/2020] [Indexed: 12/14/2022] Open
Abstract
In cancer-immunity cycle, the immune checkpoint PD1 and its ligand PDL1 act as accomplices to help tumors resist to immunity-induced apoptosis and promote tumor progression. Immunotherapy targeting PD1/PDL1 axis can effectively block its pro-tumor activity. Anti-PD1/PDL1 therapy has achieved great success in the past decade. However, only a subset of patients showed clinical responses. Most of the patients can not benefit from anti-PD1/PDL1 therapy. Furthermore, a large group of responders would develop acquired resistance after initial responses. Therefore, understanding the mechanisms of resistance is necessary for improving anti-PD1/PDL1 efficacy. Currently, researchers have identified primary resistance mechanisms which include insufficient tumor immunogenicity, disfunction of MHCs, irreversible T cell exhaustion, primary resistance to IFN-γ signaling, and immunosuppressive microenvironment. Some oncogenic signaling pathways also contribute to the primary resistance. Under the pressure applied by anti-PD1/PDL1 therapy, tumors experience immunoediting and preserve beneficial mutations, upregulate the compensatory inhibitory signaling and induce re-exhaustion of T cells, all of which may attenuate the durability of the therapy. Here we explore the underlying mechanisms in detail, review biomarkers that help identifying responders among patients and discuss the strategies that may relieve the anti-PD1/PDL1 resistance.
Collapse
Affiliation(s)
- Qingyang Lei
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Dan Wang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Kai Sun
- College of Medicine, Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
27
|
Bai R, Chen N, Liang T, Li L, Lv Z, Lv X, Cui J. Novel Frontiers of Treatment for Advanced Gastric or Gastroesophageal Junction Cancer (GC/GEJC): Will Immunotherapy Be a Future Direction? Front Oncol 2020; 10:912. [PMID: 32793461 PMCID: PMC7386300 DOI: 10.3389/fonc.2020.00912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022] Open
Abstract
Considering the limited progress of chemotherapy and targeted therapy in improving the generally disappointing outcomes of advanced gastric or gastroesophageal junction cancer (GC/GEJC), immunotherapies have been gradually developed and advanced into novel frontiers of treatment for advanced GC/GEJC. Nevertheless, the response to immunotherapy was not always satisfactory, and the emergence of resistance was unavoidable. These factors prompt the development of different combination therapies and predictive and prognostic biomarkers of efficacy to improve the outcomes of patients with advanced GC/GEJC and to overcome drug resistance. This article discusses the advances of immune monotherapy, multiple current and ongoing clinical trials of immune combination therapy, immune-related adverse events, and various biomarkers in GC/GEJC.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Naifei Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Tingting Liang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lingyu Li
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zheng Lv
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaomin Lv
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Jiuwei Cui
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Abstract
Tumor immunology is undergoing a renaissance due to the recent profound clinical successes of tumor immunotherapy. These advances have coincided with an exponential growth in the development of -omics technologies. Armed with these technologies and their associated computational and modeling toolsets, systems biologists have turned their attention to tumor immunology in an effort to understand the precise nature and consequences of interactions between tumors and the immune system. Such interactions are inherently multivariate, spanning multiple time and size scales, cell types, and organ systems, rendering systems biology approaches particularly amenable to their interrogation. While in its infancy, the field of 'Cancer Systems Immunology' has already influenced our understanding of tumor immunology and immunotherapy. As the field matures, studies will move beyond descriptive characterizations toward functional investigations of the emergent behavior that govern tumor-immune responses. Thus, Cancer Systems Immunology holds incredible promise to advance our ability to fight this disease.
Collapse
Affiliation(s)
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of MedicineStanfordUnited States
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of MedicineStanfordUnited States
- Stanford Cancer Institute, Stanford UniversityStanfordUnited States
| |
Collapse
|
29
|
Plasma-Derived Extracellular Vesicles Convey Protein Signatures that Reflect Pathophysiology in Lung and Pancreatic Adenocarcinomas. Cancers (Basel) 2020; 12:cancers12051147. [PMID: 32370304 PMCID: PMC7281335 DOI: 10.3390/cancers12051147] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/16/2022] Open
Abstract
Using a combination of mass-spectrometry and aptamer array-based proteomics, we characterized the protein features of circulating extracellular vesicles (EVs) in the context of lung (LUAD) and pancreatic ductal (PDAC) adenocarcinomas. We profiled EVs isolated from conditioned media of LUAD and PDAC cell lines to identify EV-associated protein cargoes released by these cancer cell types. Analysis of the resulting data identified LUAD and PDAC specific and pan-adenocarcinoma EV protein signatures. Bioinformatic analyses confirmed enrichment of proteins annotated to vesicle-associated processes and intracellular compartments, as well as representation of cancer hallmark functions and processes. Analysis of upstream regulator networks indicated significant enrichment of TP53, MYC, TGFB1 and KRAS-driven network effectors (p = 1.69 × 10-77-2.93 × 10-49) manifest in the adenocarcinoma sEV protein cargoes. We extended these findings by profiling the proteome of EVs isolated from lung (N = 15) and pancreatic ductal (N = 6) adenocarcinoma patient plasmas obtained at time of diagnosis, along with EVs derived from matched healthy controls (N = 21). Exploration of these proteomic data revealed abundant protein features in the plasma EVs with capacity to distinguish LUAD and PDAC cases from controls, including features yielding higher performance in the plasma EV isolates relative to unfractionated plasmas.
Collapse
|
30
|
Invrea F, Rovito R, Torchiaro E, Petti C, Isella C, Medico E. Patient-derived xenografts (PDXs) as model systems for human cancer. Curr Opin Biotechnol 2020; 63:151-156. [PMID: 32070860 DOI: 10.1016/j.copbio.2020.01.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Accepted: 01/08/2020] [Indexed: 12/13/2022]
Abstract
Patient-derived xenografts (PDXs) are obtained by transplanting fragments of a patient's tumour into immunodeficient mice. Growth and propagation of PDXs allows correlating therapeutic response in vivo with extensive, multi-dimensional molecular annotation, leading to identification of predictive biomarkers. PDXs are increasingly recognised as clinically relevant models of cancer for several reasons, of which the main is the possibility of studying the behaviour of cancer cells in a natural microenvironment, where they interact with stromal components accrued from the mouse host. PDXs maintain close similarities with the tumour of origin, in terms of tissue architecture, molecular features and response to treatments. Indeed, preclinical trials in PDXs have been shown to match and also anticipate data obtained in patients. Exploration of more complex processes like metastatic evolution and antitumour immune responses is actively pursued with PDXs, as new generations of host models emerge on the horizon.
Collapse
Affiliation(s)
- Federica Invrea
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Roberta Rovito
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Erica Torchiaro
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Consalvo Petti
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Claudio Isella
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy; University of Torino, Department of Oncology, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy
| | - Enzo Medico
- Candiolo Cancer Institute, FPO-IRCCS, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy; University of Torino, Department of Oncology, strada Prov. 142, km 3,95, 10060 Candiolo (TO), Italy.
| |
Collapse
|
31
|
Hegde PS, Chen DS. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020; 52:17-35. [PMID: 31940268 DOI: 10.1016/j.immuni.2019.12.011] [Citation(s) in RCA: 1214] [Impact Index Per Article: 242.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/01/2019] [Accepted: 12/14/2019] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a validated and critically important approach for treating patients with cancer. Given the vast research and clinical investigation efforts dedicated to advancing both endogenous and synthetic immunotherapy approaches, there is a need to focus on crucial questions and define roadblocks to the basic understanding and clinical progress. Here, we define ten key challenges facing cancer immunotherapy, which range from lack of confidence in translating pre-clinical findings to identifying optimal combinations of immune-based therapies for any given patient. Addressing these challenges will require the combined efforts of basic researchers and clinicians, and the focusing of resources to accelerate understanding of the complex interactions between cancer and the immune system and the development of improved treatment options for patients with cancer.
Collapse
|
32
|
Hou Q, Xu H. Rational Discovery of Response Biomarkers: Candidate Prognostic Factors and Biomarkers for Checkpoint Inhibitor-Based Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:143-166. [PMID: 32185710 DOI: 10.1007/978-981-15-3266-5_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Immunotherapy with checkpoint inhibitor has been successfully applied in treatment for multiple cancer types, especially for patients at advanced stage. However, response rate of this promising therapy is low, thus requiring biomarkers for precise medication to reduce the ineffective treatment. With multiple retrospective clinical studies, more and more candidate prognostic factors have been identified with possible mechanic explanation, including the basic clinical characteristics (e.g., age and gender), molecular features (e.g., PD-L1 expression and tumor mutation burden). After validation in independent patient cohorts with large sample size, several markers have been approved as companion biomarkers. However, validation and combinations of all the possible candidate biomarkers are still challenging to predict the treatment outcomes. In this chapter, we will summarize and introduce the prognostic factors and biomarkers for checkpoint inhibitor-based immunotherapy.
Collapse
Affiliation(s)
- Qianqian Hou
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Heng Xu
- Department of Laboratory Medicine, Precision Medicine Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
33
|
Li L, Wang Y, Shi W, Zhu M, Liu Z, Luo N, Zeng Y, He Y. Serial ultra-deep sequencing of circulating tumor DNA reveals the clonal evolution in non-small cell lung cancer patients treated with anti-PD1 immunotherapy. Cancer Med 2019; 8:7669-7678. [PMID: 31692284 PMCID: PMC6912064 DOI: 10.1002/cam4.2632] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/25/2019] [Accepted: 10/07/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Immune-therapy with anti-PD1 inhibitors, such as pembrolizumab, is revolutionizing the treatment of non-small cell lung cancers (NSCLC). However, identifying patients for the potential therapeutic response and predicting therapy resistance and early relapse remains a challenge. METHODS Between 2016 and 2018, 60 patients were treated with pembrolizumab, among who 12 NSCLC patients had both baseline (before treatment) and serial (on treatment) periodical circulating tumor DNA (ctDNA) samples. Those samples were sequenced on a 329 pan cancer-related gene panel. Analyses of tumor burden, blood tumor mutational burden (bTMB), maximum somatic allele frequency (MSAF), and tumor clonal structure were performed in association with clinical response. Candidate resistance mutations involved in relapse and metastases were further investigated. RESULTS ctDNA was detected and mutational profiling was performed for each patient. Those with a high baseline bTMB level showed significantly improved progression-free survival (PFS) after pembrolizumab treatment. Tumor burden and therapeutic response significantly correlated with the MSAF instead of the bTMB. Clone analysis detected tumor progression about 2-4 months ahead of computed tomography (CT) scan. One mutation in gene PTCH1 (Protein patched homolog 1) and two acquired anti-PD1 candidate resistance mutations of gene B2M (β2 microglobulin) were identified in association with distant metastasis. The evolutionary tree of a representative patient was also described. CONCLUSION This pilot study showed that MSAF could be another good indicator of therapeutic response, and clonal analysis could be clinically useful in monitoring clonal dynamics and detecting remote metastasis and early relapse.
Collapse
Affiliation(s)
- Li Li
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Yubo Wang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Mengxiao Zhu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhulin Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | - Nuo Luo
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| | | | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
34
|
Xue G, Cui ZJ, Zhou XH, Zhu YX, Chen Y, Liang FJ, Tang DN, Huang BY, Zhang HY, Hu ZH, Yuan XY, Xiong J. DNA Methylation Biomarkers Predict Objective Responses to PD-1/PD-L1 Inhibition Blockade. Front Genet 2019; 10:724. [PMID: 31475034 PMCID: PMC6707807 DOI: 10.3389/fgene.2019.00724] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) treatment could bring long-lasting clinical benefits to patients with metastatic cancer. However, only a small proportion of patients respond to PD-1/PD-L1 blockade, so predictive biomarkers are needed. Here, based on DNA methylation profiles and the objective response rates (ORRs) of PD-1/PD-L1 inhibition therapy, we identified 269 CpG sites and developed an initial CpG-based model by Lasso to predict ORRs. Notably, as measured by the area under the receiver operating characteristic curve (AUC), our model can produce better performance (AUC = 0.92) than both a model based on tumor mutational burden (TMB) (AUC = 0.77) and a previously reported TMB model (AUC = 0.71). In addition, most CpGs also have additional synergies with TMB, which can achieve a higher prediction accuracy when joined with TMB. Furthermore, we identified CpGs that are associated with TMB at the individual level. DNA methylation modules defined by protein networks, Kyoto Encylopedia of Genes and Genomes (KEGG) pathways, and ligand-receptor gene pairs are also associated with ORRs. This method suggested novel immuno-oncology targets that might be beneficial when combined with PD-1/PD-L1 blockade. Thus, DNA methylation studies might hold great potential for individualized PD1/PD-L1 blockade or combinatory therapy.
Collapse
Affiliation(s)
- Gang Xue
- SPACEnter Space Science and Technology Institute, Shenzhen, China.,College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Ze-Jia Cui
- College of Informatics, Huazhong Agricultural University, Wuhan, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xiong-Hui Zhou
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Yue-Xing Zhu
- SPACEnter Space Science and Technology Institute, Shenzhen, China
| | - Ying Chen
- SPACEnter Space Science and Technology Institute, Shenzhen, China
| | - Feng-Ji Liang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Da-Nian Tang
- Gastro-Intestinal Surgery Department, Beijing Hospital, Beijing, China
| | - Bing-Yang Huang
- Department of Cardiothoracic Surgery, Strategic Support Force Medical Center of PLA. No. 9, Beijing, China
| | - Hong-Yu Zhang
- College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Zhi-Huang Hu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xi-Yu Yuan
- Department of General Surgery, Dongguan People's Hospital affiliated to Southern Medical University, Dongguan, China
| | - Jianghui Xiong
- SPACEnter Space Science and Technology Institute, Shenzhen, China.,State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| |
Collapse
|
35
|
Cursons J, Souza-Fonseca-Guimaraes F, Foroutan M, Anderson A, Hollande F, Hediyeh-Zadeh S, Behren A, Huntington ND, Davis MJ. A Gene Signature Predicting Natural Killer Cell Infiltration and Improved Survival in Melanoma Patients. Cancer Immunol Res 2019; 7:1162-1174. [PMID: 31088844 DOI: 10.1158/2326-6066.cir-18-0500] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 01/31/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Natural killer (NK) cell activity is essential for initiating antitumor responses and may be linked to immunotherapy success. NK cells and other innate immune components could be exploitable for cancer treatment, which drives the need for tools and methods that identify therapeutic avenues. Here, we extend our gene-set scoring method singscore to investigate NK cell infiltration by applying RNA-seq analysis to samples from bulk tumors. Computational methods have been developed for the deconvolution of immune cell types within solid tumors. We have taken the NK cell gene signatures from several such tools, then curated the gene list using a comparative analysis of tumors and immune cell types. Using a gene-set scoring method to investigate RNA-seq data from The Cancer Genome Atlas (TCGA), we show that patients with metastatic cutaneous melanoma have an improved survival rate if their tumor shows evidence of NK cell infiltration. Furthermore, these survival effects are enhanced in tumors that show higher expression of genes that encode NK cell stimuli such as the cytokine IL15 Using this signature, we then examine transcriptomic data to identify tumor and stromal components that may influence the penetrance of NK cells into solid tumors. Our results provide evidence that NK cells play a role in the regulation of human tumors and highlight potential survival effects associated with increased NK cell activity. Our computational analysis identifies putative gene targets that may be of therapeutic value for boosting NK cell antitumor immunity.
Collapse
Affiliation(s)
- Joseph Cursons
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Fernando Souza-Fonseca-Guimaraes
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Momeneh Foroutan
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Clinical Pathology, The University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Ashley Anderson
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, The University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Australia
| | - Soroor Hediyeh-Zadeh
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Andreas Behren
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
| | - Nicholas D Huntington
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia.
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Melissa J Davis
- Bioinformatics Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
36
|
Saigi M, Alburquerque-Bejar JJ, Sanchez-Cespedes M. Determinants of immunological evasion and immunocheckpoint inhibition response in non-small cell lung cancer: the genetic front. Oncogene 2019; 38:5921-5932. [PMID: 31253869 DOI: 10.1038/s41388-019-0855-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/15/2019] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
The incorporation into clinical practice of immune-checkpoint inhibitors (ICIs), such as those targeting the cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and the programmed cell death 1 (PD-1) and its ligand (PD-L1), has represented a major breakthrough in non-small cell lung cancer (NSCLC) treatment, especially in cases where the cancer has no druggable genetic alterations. Despite becoming the standard of care in certain clinical settings, either alone or in combination with chemotherapy, a proportion of patients do not respond while others actually progress during treatment. Therefore, there is a clinical need to identify accurate predictive biomarkers and to develop novel therapeutic strategies based on ICIs. Although they have limitations, the current markers evaluated to select which patients will undergo ICI treatment are the levels of PD-L1 and the tumor mutational burden. In this paper we describe what is currently known about the dynamic interaction between the cancer cell and the immune system during carcinogenesis, with a particular focus on the description of the functions and gene alterations that preclude the host immunoresponse in NSCLC. We emphasize the deleterious gene alterations in components of the major histocompatibility complex (HLA-I or B2M) and of the response to IFNγ (such as JAK2) which are mutually exclusive and can affect up to one fifth of the NSCLCs. The participation of other gene alterations, such as those of common oncogenes and tumor suppressors, and of the epigenetic alterations will also be discussed, in detail. Finally, we discuss the potential use of the tumor's genetic profile to predict sensitivity to ICIs.
Collapse
Affiliation(s)
- Maria Saigi
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.,Department of Medical Oncology, Catalan Institute of Oncology (ICO), Avda Gran via, 199-203. L'Hospitalet, 08908, Barcelona, Spain
| | - Juan J Alburquerque-Bejar
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Montse Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain. .,Cancer Genomics Group, Josep Carreras Leukemia Research Institute, IJC Building, Campus ICO-Germans Trias i Pujol Ctra de Can Ruti, Camí de les Escoles s/n, Badalona, 08916, Barcelona, Spain.
| |
Collapse
|
37
|
Beane JE, Mazzilli SA, Campbell JD, Duclos G, Krysan K, Moy C, Perdomo C, Schaffer M, Liu G, Zhang S, Liu H, Vick J, Dhillon SS, Platero SJ, Dubinett SM, Stevenson C, Reid ME, Lenburg ME, Spira AE. Molecular subtyping reveals immune alterations associated with progression of bronchial premalignant lesions. Nat Commun 2019; 10:1856. [PMID: 31015447 PMCID: PMC6478943 DOI: 10.1038/s41467-019-09834-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022] Open
Abstract
Bronchial premalignant lesions (PMLs) are precursors of lung squamous cell carcinoma, but have variable outcome, and we lack tools to identify and treat PMLs at risk for progression to cancer. Here we report the identification of four molecular subtypes of PMLs with distinct differences in epithelial and immune processes based on RNA-Seq profiling of endobronchial biopsies from high-risk smokers. The Proliferative subtype is enriched with bronchial dysplasia and exhibits up-regulation of metabolic and cell cycle pathways. A Proliferative subtype-associated gene signature identifies subjects with Proliferative PMLs from normal-appearing uninvolved large airway brushings with high specificity. In progressive/persistent Proliferative lesions expression of interferon signaling and antigen processing/presentation pathways decrease and immunofluorescence indicates a depletion of innate and adaptive immune cells compared with regressive lesions. Molecular biomarkers measured in PMLs or the uninvolved airway can enhance histopathological grading and suggest immunoprevention strategies for intercepting the progression of PMLs to lung cancer.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biopsy
- Bronchi/diagnostic imaging
- Bronchi/immunology
- Bronchi/pathology
- Bronchoscopy
- Carcinoma, Bronchogenic/genetics
- Carcinoma, Bronchogenic/immunology
- Carcinoma, Bronchogenic/pathology
- Carcinoma, Bronchogenic/prevention & control
- Cohort Studies
- Datasets as Topic
- Disease Progression
- Early Detection of Cancer/methods
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/immunology
- Gene Regulatory Networks/genetics
- Gene Regulatory Networks/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Cellular/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Mass Screening/methods
- Middle Aged
- Precancerous Conditions/diagnostic imaging
- Precancerous Conditions/genetics
- Precancerous Conditions/immunology
- Precancerous Conditions/pathology
- RNA, Messenger/genetics
- Respiratory Mucosa/cytology
- Respiratory Mucosa/diagnostic imaging
- Respiratory Mucosa/immunology
- Respiratory Mucosa/pathology
- Sequence Analysis, RNA
- T-Lymphocytes/immunology
- Tomography, X-Ray Computed
- Up-Regulation
Collapse
Affiliation(s)
| | | | | | - Grant Duclos
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Kostyantyn Krysan
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | | | | | - Gang Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sherry Zhang
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hanqiao Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Jessica Vick
- Boston University School of Medicine, Boston, MA, 02118, USA
| | | | | | - Steven M Dubinett
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | - Mary E Reid
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Marc E Lenburg
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Avrum E Spira
- Boston University School of Medicine, Boston, MA, 02118, USA
- Johnson and Johnson Innovation, Cambridge, MA, 02142, USA
| |
Collapse
|
38
|
Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy Not Work for All Patients? Am Soc Clin Oncol Educ Book 2019; 39:147-164. [PMID: 31099674 DOI: 10.1200/edbk_240837] [Citation(s) in RCA: 451] [Impact Index Per Article: 75.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
The emergence of immune checkpoint blockade therapies over the last decade has transformed cancer treatment in a wide range of tumor types. Unprecedented and durable clinical responses in difficult-to-treat cancer histologies have been observed. However, despite these promising long-term responses, the majority of patients fail to respond to immune checkpoint blockade, demonstrating primary resistance. Additionally, many of those who initially respond to treatment eventually experience relapse secondary to acquired resistance. Both primary and acquired resistance are a result of complex and constantly evolving interactions between cancer cells and the immune system. Many mechanisms of resistance have been characterized to date, and more continue to be uncovered. By elucidating and targeting mechanisms of resistance, treatments can be tailored to improve clinical outcomes. This review will discuss the landscape of immune checkpoint blockade response data, different resistance mechanisms, and potential therapeutic strategies to overcome resistance.
Collapse
Affiliation(s)
- Charlene M Fares
- 1 Department of Medicine, Division of Hematology/Oncology, University of California, Los Angeles, Los Angeles, CA
| | | | - Charles G Drake
- 3 Department of Medicine, Division of Hematology/Oncology, Columbia University Medical Center, New York, NY
| | - James P Allison
- 4 Department of Immunology, Division of Basic Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Siwen Hu-Lieskovan
- 5 Division of Hematology and Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| |
Collapse
|
39
|
Pitt JJ, Riester M, Zheng Y, Yoshimatsu TF, Sanni A, Oluwasola O, Veloso A, Labrot E, Wang S, Odetunde A, Ademola A, Okedere B, Mahan S, Leary R, Macomber M, Ajani M, Johnson RS, Fitzgerald D, Grundstad AJ, Tuteja JH, Khramtsova G, Zhang J, Sveen E, Hwang B, Clayton W, Nkwodimmah C, Famooto B, Obasi E, Aderoju V, Oludara M, Omodele F, Akinyele O, Adeoye A, Ogundiran T, Babalola C, MacIsaac K, Popoola A, Morrissey MP, Chen LS, Wang J, Olopade CO, Falusi AG, Winckler W, Haase K, Van Loo P, Obafunwa J, Papoutsakis D, Ojengbede O, Weber B, Ibrahim N, White KP, Huo D, Olopade OI, Barretina J. Characterization of Nigerian breast cancer reveals prevalent homologous recombination deficiency and aggressive molecular features. Nat Commun 2018; 9:4181. [PMID: 30327465 PMCID: PMC6191428 DOI: 10.1038/s41467-018-06616-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 09/13/2018] [Indexed: 02/08/2023] Open
Abstract
Racial/ethnic disparities in breast cancer mortality continue to widen but genomic studies rarely interrogate breast cancer in diverse populations. Through genome, exome, and RNA sequencing, we examined the molecular features of breast cancers using 194 patients from Nigeria and 1037 patients from The Cancer Genome Atlas (TCGA). Relative to Black and White cohorts in TCGA, Nigerian HR + /HER2 - tumors are characterized by increased homologous recombination deficiency signature, pervasive TP53 mutations, and greater structural variation-indicating aggressive biology. GATA3 mutations are also more frequent in Nigerians regardless of subtype. Higher proportions of APOBEC-mediated substitutions strongly associate with PIK3CA and CDH1 mutations, which are underrepresented in Nigerians and Blacks. PLK2, KDM6A, and B2M are also identified as previously unreported significantly mutated genes in breast cancer. This dataset provides novel insights into potential molecular mechanisms underlying outcome disparities and lay a foundation for deployment of precision therapeutics in underserved populations.
Collapse
Affiliation(s)
- Jason J Pitt
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Markus Riester
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Toshio F Yoshimatsu
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Ayodele Sanni
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | - Artur Veloso
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Emma Labrot
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Shengfeng Wang
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, 100191, China
| | - Abayomi Odetunde
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Adeyinka Ademola
- Department of Surgery, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Babajide Okedere
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Scott Mahan
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Rebecca Leary
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Maura Macomber
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Mustapha Ajani
- Department of Pathology, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Ryan S Johnson
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Dominic Fitzgerald
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - A Jason Grundstad
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jigyasa H Tuteja
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
| | - Galina Khramtsova
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jing Zhang
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Elisabeth Sveen
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Bryce Hwang
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Wendy Clayton
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | - Bisola Famooto
- Department of Surgery, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Esther Obasi
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Victor Aderoju
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Mobolaji Oludara
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Folusho Omodele
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Odunayo Akinyele
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Adewunmi Adeoye
- Department of Pathology, University of Ibadan, Ibadan, Oyo, Nigeria
| | | | - Chinedum Babalola
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
- Department of Pharmaceutical Chemistry, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Kenzie MacIsaac
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Abiodun Popoola
- Oncology Unit, Department of Radiology, Lagos State University, Ikeja, Lagos, Nigeria
| | | | - Lin S Chen
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Jiebiao Wang
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Christopher O Olopade
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Adeyinka G Falusi
- Institute for Advanced Medical Research and Training, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Wendy Winckler
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Kerstin Haase
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Peter Van Loo
- The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Department of Human Genetics, University of Leuven, Oude Markt 13, Leuven, 3000, Belgium
| | - John Obafunwa
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | | | - Oladosu Ojengbede
- Centre for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Oyo, Nigeria
| | - Barbara Weber
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA
| | - Nasiru Ibrahim
- Department of Surgery, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Kevin P White
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
- Tempus Labs Inc., Chicago, IL, USA.
| | - Dezheng Huo
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
- Department of Public Health Sciences, University of Chicago, Chicago, IL 60637, USA.
| | - Olufunmilayo I Olopade
- Institute for Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA.
- Center for Clinical Cancer Genetics & Global Health, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | - Jordi Barretina
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139, USA.
- Girona Biomedical Research Institute (IDIBGI), Girona, 17007, Spain.
| |
Collapse
|
40
|
Deep analysis of acquired resistance to FGFR1 inhibitor identifies MET and AKT activation and an expansion of AKT1 mutant cells. Oncotarget 2018; 9:31549-31558. [PMID: 30140389 PMCID: PMC6101141 DOI: 10.18632/oncotarget.25862] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 07/16/2018] [Indexed: 11/25/2022] Open
Abstract
The development of acquired resistance (AR) to tyrosine kinase inhibitors (TKIs) of FGFR1 activation is currently not well understood. To gain a deeper insight into this matter in lung cancer, we used the FGFR1-amplified DMS114 cell line and generated multiple clones with AR to an FGFR1-TKI. We molecularly scrutinized the resistant cells, using whole-exome sequencing, RNA sequencing and global DNA methylation analysis. Our results show a de novo activation of AKT and ERK, and a reactivation of mTOR. Furthermore, the resistant cells exhibited strong upregulation and activation of MET, indicating crosstalk between the FGFR1 and MET axes. The resistant cells also underwent a global decrease in promoter hypermethylation of the CpG islands. Finally, we observed clonal expansion of a pre-existing change in AKT1, leading to S266L substitution, within the kinase domain of AKT. Our results demonstrate that AR to FGFR1-TKI involves deep molecular changes that promote the activation of MET and AKT, coupled with common gene expression and DNA methylation profiles. The expansion of a substitution at AKT1 was the only shared genetic change, and this may have contributed to the AR.
Collapse
|
41
|
Saigi M, Alburquerque-Bejar JJ, Mc Leer-Florin A, Pereira C, Pros E, Romero OA, Baixeras N, Esteve-Codina A, Nadal E, Brambilla E, Sanchez-Cespedes M. MET-Oncogenic and JAK2-Inactivating Alterations Are Independent Factors That Affect Regulation of PD-L1 Expression in Lung Cancer. Clin Cancer Res 2018; 24:4579-4587. [PMID: 29898990 DOI: 10.1158/1078-0432.ccr-18-0267] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/10/2018] [Accepted: 06/06/2018] [Indexed: 11/16/2022]
Abstract
Purpose: The blockade of immune checkpoints such as PD-L1 and PD-1 is being exploited therapeutically in several types of malignancies. Here, we aimed to understand the contribution of the genetics of lung cancer to the ability of tumor cells to escape immunosurveillance checkpoints.Experimental Design: More than 150 primary non-small cell lung cancers, including pulmonary sarcomatoid carcinomas, were tested for levels of the HLA-I complex, PD-L1, tumor-infiltrating CD8+ lymphocytes, and alterations in main lung cancer genes. Correlations were validated in cancer cell lines using appropriate treatments to activate or inhibit selected pathways. We also performed RNA sequencing to assess changes in gene expression after these treatments.Results:MET-oncogenic activation tended to associate with positive PD-L1 immunostaining, whereas STK11 mutations were correlated with negative immunostaining. In MET-altered cancer cells, MET triggered a transcriptional increase of PD-L1 that was independent of the IFNγ-mediated JAK/STAT pathway. The activation of MET also upregulated other immunosuppressive genes (PDCD1LG2 and SOCS1) and transcripts involved in angiogenesis (VEGFA and NRP1) and in cell proliferation. We also report recurrent inactivating mutations in JAK2 that co-occur with alterations in MET and STK11, which prevented the induction of immunoresponse-related genes following treatment with IFNγ.Conclusions: We show that MET activation promotes the expression of several negative checkpoint regulators of the immunoresponse, including PD-L1. In addition, we report inactivation of JAK2 in lung cancer cells that prevented the response to IFNγ. These alterations are likely to facilitate tumor growth by enabling immune tolerance and may affect the response to immune checkpoint inhibitors. Clin Cancer Res; 24(18); 4579-87. ©2018 AACR.
Collapse
Affiliation(s)
- Maria Saigi
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Juan J Alburquerque-Bejar
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Anne Mc Leer-Florin
- Département d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble and Université Grenoble Alpes, Grenoble, France
| | - Carolina Pereira
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Eva Pros
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Octavio A Romero
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Baixeras
- Pathology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain.,Clinical Research in Solid Tumors (CReST) Group, OncoBell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Elisabeth Brambilla
- Département d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble and Université Grenoble Alpes, Grenoble, France
| | - Montse Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
42
|
Cai L, Michelakos T, Yamada T, Fan S, Wang X, Schwab JH, Ferrone CR, Ferrone S. Defective HLA class I antigen processing machinery in cancer. Cancer Immunol Immunother 2018; 67:999-1009. [PMID: 29487978 PMCID: PMC8697037 DOI: 10.1007/s00262-018-2131-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 02/06/2018] [Indexed: 11/26/2022]
Abstract
Malignant transformation of cells is frequently associated with defective HLA class I antigen processing machinery (APM) component expression. This abnormality may have functional relevance, since it may have a negative impact on tumor cell recognition by cognate T cells. Furthermore, HLA class I APM abnormalities appear to have clinical significance, since they are associated with poor prognosis in several malignant diseases and may play a role in the resistance to immune checkpoint inhibitor-based immunotherapy. In this paper, we have reviewed the literature describing abnormalities in HLA class I APM component expression in many types of cancer. These abnormalities have been reported in all types of cancer analyzed with a frequency ranging between a minimum of 35.8% in renal cancer and a maximum of 87.9% in thyroid cancer for HLA class I heavy chains. In addition, we have described the molecular mechanisms underlying defects in HLA class I APM component expression and function by malignant cells. Lastly, we have discussed the clinical significance of HLA class I APM component abnormalities in malignant tumors.
Collapse
Affiliation(s)
- Lei Cai
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Hepatobiliary, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Theodoros Michelakos
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Teppei Yamada
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Song Fan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Xinhui Wang
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph H Schwab
- Department of Orthopedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
43
|
De-novo and acquired resistance to immune checkpoint targeting. Lancet Oncol 2017; 18:e731-e741. [PMID: 29208439 DOI: 10.1016/s1470-2045(17)30607-1] [Citation(s) in RCA: 511] [Impact Index Per Article: 63.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 07/21/2017] [Accepted: 07/24/2017] [Indexed: 12/19/2022]
Abstract
Use of immune checkpoint inhibitors targeting the programmed cell death protein-1/programmed cell death-ligand 1 and cytotoxic T lymphocyte-associated protein-4 axes has yielded impressive results in some clinical trials. However, only a subset of patients initially respond to these inhibitors, and increasing clinical evidence indicates that a substantial proportion of initial responders ultimately relapse with lethal, drug-resistant disease months or years later. Studies that have used massively parallel sequencing have shed light on the rich functional landscape of mutations that endow tumour cells with the ability to evade T-cell-mediated immunosurveillance. Cancer genomes bear signatures of clonal evolution and selection, particularly implicating acquired defects in interferon receptor signalling and antigen presentation. In this Review, we discuss the biological processes that operate in the formation of so-called immunoresistant niches, and describe the latest progress in the development of combination strategies to reinstate immunosurveillance in immune-refractory tumours.
Collapse
|
44
|
Gettinger S, Choi J, Hastings K, Truini A, Datar I, Sowell R, Wurtz A, Dong W, Cai G, Melnick MA, Du VY, Schlessinger J, Goldberg SB, Chiang A, Sanmamed MF, Melero I, Agorreta J, Montuenga LM, Lifton R, Ferrone S, Kavathas P, Rimm DL, Kaech SM, Schalper K, Herbst RS, Politi K. Impaired HLA Class I Antigen Processing and Presentation as a Mechanism of Acquired Resistance to Immune Checkpoint Inhibitors in Lung Cancer. Cancer Discov 2017; 7:1420-1435. [PMID: 29025772 DOI: 10.1158/2159-8290.cd-17-0593] [Citation(s) in RCA: 513] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/25/2017] [Accepted: 09/29/2017] [Indexed: 12/11/2022]
Abstract
Mechanisms of acquired resistance to immune checkpoint inhibitors (ICI) are poorly understood. We leveraged a collection of 14 ICI-resistant lung cancer samples to investigate whether alterations in genes encoding HLA Class I antigen processing and presentation machinery (APM) components or interferon signaling play a role in acquired resistance to PD-1 or PD-L1 antagonistic antibodies. Recurrent mutations or copy-number changes were not detected in our cohort. In one case, we found acquired homozygous loss of B2M that caused lack of cell-surface HLA Class I expression in the tumor and a matched patient-derived xenograft (PDX). Downregulation of B2M was also found in two additional PDXs established from ICI-resistant tumors. CRISPR-mediated knockout of B2m in an immunocompetent lung cancer mouse model conferred resistance to PD-1 blockade in vivo, proving its role in resistance to ICIs. These results indicate that HLA Class I APM disruption can mediate escape from ICIs in lung cancer.Significance: As programmed death 1 axis inhibitors are becoming more established in standard treatment algorithms for diverse malignancies, acquired resistance to these therapies is increasingly being encountered. Here, we found that defective antigen processing and presentation can serve as a mechanism of such resistance in lung cancer. Cancer Discov; 7(12); 1420-35. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1355.
Collapse
Affiliation(s)
- Scott Gettinger
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut. .,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Jungmin Choi
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Katherine Hastings
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Anna Truini
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Ila Datar
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Ryan Sowell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Anna Wurtz
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Weilai Dong
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Guoping Cai
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Mary Ann Melnick
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Victor Y Du
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Joseph Schlessinger
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Sarah B Goldberg
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut.,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Anne Chiang
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut.,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Miguel F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Ignacio Melero
- CIMA and Clinica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en red de Oncología CIBERONC, Madrid, Spain
| | - Jackeline Agorreta
- CIMA and Clinica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en red de Oncología CIBERONC, Madrid, Spain
| | - Luis M Montuenga
- CIMA and Clinica Universidad de Navarra, Pamplona, Spain.,Centro de Investigación Biomédica en red de Oncología CIBERONC, Madrid, Spain
| | - Richard Lifton
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Paula Kavathas
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut.,Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - David L Rimm
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Susan M Kaech
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut
| | - Kurt Schalper
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut.,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Roy S Herbst
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut.,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut
| | - Katerina Politi
- Department of Medicine (Section of Medical Oncology), Yale University School of Medicine, New Haven, Connecticut. .,Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut.,Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|