51
|
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: 4.3] [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
|
52
|
Immune-Checkpoint Inhibitors in B-Cell Lymphoma. Cancers (Basel) 2021; 13:cancers13020214. [PMID: 33430146 PMCID: PMC7827333 DOI: 10.3390/cancers13020214] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/16/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Immune-based treatment strategies, which include immune checkpoint inhibition, have recently become a new frontier for the treatment of B-cell-derived lymphoma. Whereas checkpoint inhibition has given oncologists and patients hope in specific lymphoma subtypes like Hodgkin lymphoma, other entities do not benefit from such promising agents. Understanding the factors that determine the efficacy and safety of checkpoint inhibition in different lymphoma subtypes can lead to improved therapeutic strategies, including combinations with various chemotherapies, biologics and/or different immunologic agents with manageable safety profiles. Abstract For years, immunotherapy has been considered a viable and attractive treatment option for patients with cancer. Among the immunotherapy arsenal, the targeting of intratumoral immune cells by immune-checkpoint inhibitory agents has recently revolutionised the treatment of several subtypes of tumours. These approaches, aimed at restoring an effective antitumour immunity, rapidly reached the market thanks to the simultaneous identification of inhibitory signals that dampen an effective antitumor response in a large variety of neoplastic cells and the clinical development of monoclonal antibodies targeting checkpoint receptors. Leading therapies in solid tumours are mainly focused on the cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD-1) pathways. These approaches have found a promising testing ground in both Hodgkin lymphoma and non-Hodgkin lymphoma, mainly because, in these diseases, the malignant cells interact with the immune system and commonly provide signals that regulate immune function. Although several trials have already demonstrated evidence of therapeutic activity with some checkpoint inhibitors in lymphoma, many of the immunologic lessons learned from solid tumours may not directly translate to lymphoid malignancies. In this sense, the mechanisms of effective antitumor responses are different between the different lymphoma subtypes, while the reasons for this substantial difference remain partially unknown. This review will discuss the current advances of immune-checkpoint blockade therapies in B-cell lymphoma and build a projection of how the field may evolve in the near future. In particular, we will analyse the current strategies being evaluated both preclinically and clinically, with the aim of fostering the use of immune-checkpoint inhibitors in lymphoma, including combination approaches with chemotherapeutics, biological agents and/or different immunologic therapies.
Collapse
|
53
|
Heinrich B, Brown ZJ, Diggs LP, Vormehr M, Ma C, Subramanyam V, Rosato U, Ruf B, Walz JS, McVey JC, Wabitsch SF, Fu Q, Yu SJ, Zhang Q, Lai CW, Sahin U, Greten TF. Steatohepatitis Impairs T-cell-Directed Immunotherapies Against Liver Tumors in Mice. Gastroenterology 2021; 160:331-345.e6. [PMID: 33010248 PMCID: PMC7755834 DOI: 10.1053/j.gastro.2020.09.031] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/23/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic steatohepatitis causes loss of hepatic CD4+ T cells and promotes tumor growth. The liver is the most common site of distant metastases from a variety of malignancies, many of which respond to immunotherapy. We investigated the effects of steatohepatitis on the efficacy of immunotherapeutic agents against liver tumors in mice. METHODS Steatohepatitis was induced by feeding C57BL/6NCrl or BALB/c AnNCr mice a methionine and choline-deficient diet or a choline-deficient l-amino acid-defined diet. Mice were given intrahepatic or subcutaneous injections of B16 melanoma and CT26 colon cancer cells, followed by intravenous injections of M30-RNA vaccine (M30) or intraperitoneal injections of an antibody against OX40 (aOX40) on days 3, 7, and 10 after injection of the tumor cells. We measured tumor growth and analyzed immune cells in tumor tissues by flow cytometry. Mice were given N-acetylcysteine to prevent loss of CD4+ T cells from liver. RESULTS Administration of M30 and aOX40 inhibited growth of tumors from intrahepatic injections of B16 or CT26 cells in mice on regular diet. However, M30 and/or aOX40 did not slow growth of liver tumors from B16 or CT26 cells in mice with diet-induced steatohepatitis (methionine and choline-deficient diet or choline-deficient l-amino acid-defined diet). Steatohepatitis did not affect the ability of M30 to slow growth of subcutaneous B16 tumors. In mice with steatohepatitis given N-acetylcysteine, which prevents loss of CD4+ T cells, M30 and aOX40 were able slow growth of hepatic tumors. Flow cytometry analysis of liver tumors revealed reduced CD4+ T cells and effector memory cells in mice with vs without steatohepatitis. CONCLUSIONS Steatohepatitis reduces the abilities of immunotherapeutic agents, such as M30 and aOX40, to inhibit tumor liver growth by reducing tumor infiltration by CD4+ T cells and effector memory cells. N-acetylcysteine restores T-cell numbers in tumors and increases the ability of M30 and aOX40 to slow tumor growth in mice.
Collapse
Affiliation(s)
- Bernd Heinrich
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zachary J. Brown
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Rutgers Robert Wood Johnson Medical School, Department of Surgery, New Brunswick, NJ
| | - Laurence P. Diggs
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mathias Vormehr
- Biopharmaceutical New Technologies (BioNTech) Corporation, An der Goldgrube 12, 55131 Mainz, Germany.,University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Chi Ma
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Varun Subramanyam
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Umberto Rosato
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Benjamin Ruf
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Juliane S. Walz
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), University Hospital Tübingen, Germany
| | - John C. McVey
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Simon F. Wabitsch
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiong Fu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Su Jong Yu
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Qianfei Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chunwei W. Lai
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Disease, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ugur Sahin
- Biopharmaceutical New Technologies (BioNTech) Corporation, An der Goldgrube 12, 55131 Mainz, Germany.,University Medical Center of the Johannes Gutenberg University, Langenbeckstraße 1, 55131 Mainz, Germany.,TRON – Translational Oncology at the University Medical Center of Johannes Gutenberg University gGmbH, Freiligrathstraße 12, 55131 Mainz, Germany
| | - Tim F. Greten
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.,NCI CCR Liver Cancer Program.,Correspondence: Tim F. Greten, MD, Thoracic and GI Malignancy Section, TGMB NIH/NCI/CCR, Building 10, Rm 2B28B, 9000 Rockville Pike, Bethesda MD 20892, USA, Telephone: +1 (240) 760 6114,
| |
Collapse
|
54
|
Green SE, McCusker MG, Mehra R. Emerging immune checkpoint inhibitors for the treatment of head and neck cancers. Expert Opin Emerg Drugs 2020; 25:501-514. [PMID: 33196319 DOI: 10.1080/14728214.2020.1852215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: The benefits of immune checkpoint inhibitors (ICIs) in recurrent and/or metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) have been demonstrated through multiple studies to improve overall survival (OS) with decreased side effects when compared to the standard of care (SOC) treatment regimens in place for decades, leading to the approval of two ICIs, nivolumab and pembrolizumab. There has been a subsequent influx in the development of novel immunotherapy agents for the treatment of HNSCC. Areas covered: Data for anti-programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte associated protein 4 (CTLA-4) antibodies in treatment of R/M HNSCC will be reviewed. Emerging immune checkpoint inhibitors as well as combined therapies in HNSCC will be discussed. The role of predictive biomarkers, HPV-status, PD-L1 expression, and challenges related to treating patients with ICIs will be summarized. Expert opinion: A shift toward ICIs as SOC for the treatment of R/M HNSCC will continue as emerging immune checkpoints and combination therapies are evaluated. Response rates are variable in this patient population underlying the importance of identifying predictive biomarkers to aid in patient selection for ICI treatment.
Collapse
Affiliation(s)
- Sarah E Green
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| | - Michael G McCusker
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| | - Ranee Mehra
- University of Maryland Medical Center, Greenebaum Comprehensive Cancer Center , Baltimore, MD, USA
| |
Collapse
|
55
|
Decreased immune response in monkeys administered a human T-effector cell agonist (OX40) antibody. Toxicol Appl Pharmacol 2020; 409:115285. [PMID: 33069749 DOI: 10.1016/j.taap.2020.115285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/25/2020] [Accepted: 10/12/2020] [Indexed: 11/21/2022]
Abstract
The OX40 receptor plays a crucial co-stimulatory role in T effector cell survival, expansion, cytokine production, and cytotoxicity to tumor cells; therefore, OX40 agonists are being evaluated as anti-cancer immunotherapies, especially in combination with checkpoint inhibitors. To support clinical development of BMS-986178 (an OX40 agonist antibody), two repeat-dose toxicity studies were conducted in cynomolgus monkeys. In the first study, BMS-986178 was administered intravenously (IV) once weekly for one month at doses from 30 to 120 mg/kg. BMS-986178 was well tolerated; surprisingly, immune function was suppressed rather than increased based on pharmacodynamic (PD) and flow cytometry readouts (e.g. T-cell dependent antibody response [TDAR]). To determine whether immune suppression was due to a bi-phasic response, a follow-up study was conducted at lower doses (1 and 10 mg/kg). Although receptor engagement was confirmed, immune function was still suppressed at both doses. In addition, treatment-emergent anti-drug antibodies (ADAs) at 1 mg/kg resulted in hypersensitivity reactions and reduced BMS-986178 exposure after repeated dosing, which precluded a full PD assessment at this dose. In conclusion, BMS-986178 was clinically well-tolerated by monkeys at weekly IV doses from 10 to 120 mg/kg (AUC[0-168] ≤ 712,000 μg●h/mL). However, despite target engagement, PD assays and other immune endpoints demonstrated immune suppression, not stimulation. Due to the inverted immune response at higher doses and the onset of ADAs, additional repeat-dose toxicity studies of BMS-986178 in monkeys (that would typically be required to support Phase 3 clinical trials and registration) would not add value for human safety assessment.
Collapse
|
56
|
Yin P, Gui L, Wang C, Yan J, Liu M, Ji L, Wang Y, Ma B, Gao WQ. Targeted Delivery of CXCL9 and OX40L by Mesenchymal Stem Cells Elicits Potent Antitumor Immunity. Mol Ther 2020; 28:2553-2563. [PMID: 32827461 DOI: 10.1016/j.ymthe.2020.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/21/2020] [Accepted: 08/05/2020] [Indexed: 12/31/2022] Open
Abstract
Major obstacles in immunotherapies include toxicities associated with systemic administration of therapeutic agents, as well as low tumor lymphocyte infiltration that hampers the efficacies. In this study, we report a mesenchymal stem cell (MSC)-based immunotherapeutic strategy in which MSCs specifically deliver T/natural killer (NK) cell-targeting chemokine CXCL9 and immunostimulatory factor OX40 ligand (OX40L)/tumor necrosis factor superfamily member 4 (TNFSF4) to tumor sites in syngeneic subcutaneous and azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced spontaneous colon cancer mouse models. This approach generated potent local antitumor immunity by increasing the ratios of tumor-infiltrating CD8+ T and NK cells and production of antitumor cytokines and cytolytic proteins in the tumor microenvironment. Moreover, it improved the efficacy of programmed death-1 (PD-1) blockade in a syngeneic mouse model and significantly suppressed the growth of major histocompatibility complex class I (MHC class I)-deficient tumors. Our MSC-based immunotherapeutic strategy simultaneously recruits and activates immune effector cells at the tumor site, thus overcoming the problems with toxicities of systemic therapeutic agents and low lymphocyte infiltration of solid tumors.
Collapse
Affiliation(s)
- Pan Yin
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liming Gui
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Caihong Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingjing Yan
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Min Liu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lu Ji
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China
| | - You Wang
- Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Bin Ma
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China; Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
57
|
Bilusic M, Girardi D, Zhou Y, Jung K, Pei J, Slifker M, Chen Q, Meerzaman D, Alpaugh K, Young D, Flieder D, Gray P, Plimack E. Molecular Profiling of Exceptional Responders to Cancer Therapy. Oncologist 2020; 26:186-195. [PMID: 33210795 DOI: 10.1002/onco.13600] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 11/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The vast majority of metastatic cancers cannot be cured. Palliative treatment may relieve disease symptoms by stopping or slowing cancer growth and may prolong patients' lives, but almost all patients will inevitably develop disease progression after initial response. However, for reasons that are not fully understood, a very few patients will have extraordinary durable responses to standard anticancer treatments. MATERIALS AND METHODS We analyzed exceptional responders treated at Fox Chase Cancer Center between September 2009 and November 2017. An exceptional response was defined as a complete response lasting more than 1 year or a partial response or stable disease for more than 2 years. Tumor samples were analyzed using an Ambry Genetics test kit with a 142-gene panel. Messenger RNA expression was evaluated using NanoString's nCounter PanCancer Pathways Panel and Immune Profiling Panel and compared with matched controls for gender, age, and cancer type. RESULTS Twenty-six exceptional responders with metastatic bladder, kidney, breast, lung, ovarian, uterine, and colon cancers were enrolled. Mutations were identified in 45 genes. The most common mutation was an EPHA5 nonsynonymous mutation detected in 87.5% of patients. Mutations in DNA damage repair pathway genes were also frequent, suggesting increased genome instability. We also found varying expression of 73 genes in the Pathways panel and 85 genes in the Immune Profiling panel, many of them responsible for improvement in tumor recognition and antitumor immune response. CONCLUSIONS The genomic instability detected in our exceptional responders, plus treatment with DNA damage compounds combined with favorable anticancer immunity, may have contributed to exceptional responses to standard anticancer therapies in the patients studied. IMPLICATIONS FOR PRACTICE With recent advances in the treatment of cancer, there is increased emphasis on the importance of identifying molecular markers to predict treatment outcomes, thereby allowing precision oncology. In this study, it was hypothesized that there is a "specific biologic signature" in the biology of the cancer in long-term survivors that allows sensitivity to systemic therapy and durability of response. Results showed that DNA damage repair pathway alterations, combined with favorable anticancer immunity, may have contributed to exceptional responses. It is very likely that an in-depth examination of outlier responses will become a standard component of drug development in the future.
Collapse
Affiliation(s)
- Marijo Bilusic
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA.,Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel Girardi
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Yan Zhou
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Kyungsuk Jung
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - Jianming Pei
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | | - Qingrong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Daoud Meerzaman
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Denise Young
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | | | | | | |
Collapse
|
58
|
Faghih Z, Taherifard E, Daneshmand A, Talei A, Erfani N. OX40 genetic variations in patients with breast cancer: a case-control study. Br J Biomed Sci 2020; 78:44-46. [PMID: 32921275 DOI: 10.1080/09674845.2020.1776587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Z Faghih
- Shiraz Institute for Cancer Research
| | | | | | - A Talei
- Breast Disease Research Center
| | - N Erfani
- Shiraz Institute for Cancer Research.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences , Shiraz, Iran
| |
Collapse
|
59
|
He S, Xu J, Wu J. The emerging role of co-stimulatory molecules and their agonistic mAb-based combination therapies in melanoma. Int Immunopharmacol 2020; 89:107097. [PMID: 33091814 DOI: 10.1016/j.intimp.2020.107097] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/09/2020] [Accepted: 10/09/2020] [Indexed: 12/26/2022]
Abstract
Although anti-PD-1/L1 and anti-CTLA-4 antibodies, the validated immune checkpoint blockades, can elicit durable long-lasting antitumor immunity and improve the clinical outcomes of melanoma treatment, there are still a fraction of patients who did not receive therapeutic benefits as expected. In addition to findings of blocking the co-inhibitory pathways, the preclinical and clinical evidence suggests that triggering the co-stimulatory pathways through agonists such as CD137, OX40, CD40, GITR and CD27 may be a rational next step for melanoma therapy. In this review, we discuss the progress of studies on these co-stimulatory molecules in terms of their promising therapeutic effects and underlying antitumor mechanisms, and provide a review of the possible combinations that orchestrate the interplay of co-stimulatory agonistic mAbs and other therapies for treating melanoma, including inhibitory immune checkpoint mAbs, adoptive T cell therapy, chemotherapy and radiotherapy. We also briefly present the limitations and challenges involved in these co-stimulatory agonistic mAb-based combination strategies for melanoma patients.
Collapse
Affiliation(s)
- Shan He
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jinhua Xu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Jinfeng Wu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai 200040, PR China.
| |
Collapse
|
60
|
Mo S, Gu L, Xu W, Liu J, Ding D, Wang Z, Yang J, Kong L, Zhao Y. Bifunctional macromolecule activating both OX40 and interferon-α signaling displays potent therapeutic effects in mouse HBV and tumor models. Int Immunopharmacol 2020; 89:107099. [PMID: 33091819 DOI: 10.1016/j.intimp.2020.107099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/10/2020] [Accepted: 10/10/2020] [Indexed: 11/16/2022]
Abstract
Combinatory enhancement of innate and adaptive immune responses is a promising strategy in immunotherapeutic drug development. Bifunctional macromolecules that simultaneously target two mechanisms may provide additional advantages over the combination of targeting two single pathways. Interferon alpha (IFNα) has been used clinically against viral infection such as the chronic infection of hepatitis B virus (CHB) as well as some types of cancers. OX40 is a costimulatory immune checkpoint molecule involved in the activation of T lymphocytes. To test whether simultaneously activating IFNα and OX40 signaling pathway could produce a synergistic therapeutic effect on CHB and tumors, we designed a bifunctional fusion protein composed of a mouse OX40 agonistic monoclonal antibody (OX86) and a mouse IFNα4, joined by a flexible (GGGGS)3 linker. This fusion protein, termed OX86-IFN, can activate both IFNα and OX40. We demonstrated that OX86-IFN could effectively activate T lymphocytes in the peripheral blood of mice. Furthermore, we showed that OX86-IFN had superior therapeutic effect to monotherapies in HBV hydrodynamic transfection and syngeneic tumor models. Collectively, our data suggests that simultaneously targeting interferon and OX40 signaling pathways by bifunctional molecule OX86-IFN elicits potent antiviral and antitumor activities, which could provide a new strategy in developing therapeutic agents against viral infection and tumors.
Collapse
Affiliation(s)
- Shifu Mo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, PR China; Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Liyun Gu
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Wei Xu
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Jia Liu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, PR China
| | - Dong Ding
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Zhichao Wang
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Jie Yang
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China
| | - Lingdong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 163 Xianlin Avenue, Nanjing, Jiangsu 210023, PR China.
| | - Yong Zhao
- Nanjing U-Mab Biopharma Co., Ltd, 699-8 Xuanwu Avenue, Nanjing, Jiangsu 210042, PR China.
| |
Collapse
|
61
|
Goggi JL, Tan YX, Hartimath SV, Jieu B, Hwang YY, Jiang L, Boominathan R, Cheng P, Yuen TY, Chin HX, Tang JR, Larbi A, Chacko AM, Renia L, Johannes C, Robins EG. Granzyme B PET Imaging of Immune Checkpoint Inhibitor Combinations in Colon Cancer Phenotypes. Mol Imaging Biol 2020; 22:1392-1402. [PMID: 32705455 PMCID: PMC7497445 DOI: 10.1007/s11307-020-01519-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune checkpoint inhibitor (ICI) monotherapy and combination regimens are being actively pursued as strategies to improve durable response rates in cancer patients. However, the biology surrounding combination therapies is not well understood and may increase the likelihood of immune-mediated adverse events. Accurate stratification of ICI response by non-invasive PET imaging may help ensure safe therapy management across a wide number of cancer phenotypes. PROCEDURES We have assessed the ability of a fluorine-labelled peptide, [18F]AlF-mNOTA-GZP, targeting granzyme B, to stratify ICI response in two syngeneic models of colon cancer, CT26 and MC38. In vivo tumour uptake of [18F]AlF-mNOTA-GZP following ICI monotherapy, or in combination with PD-1 was characterised and correlated with changes in tumour-associated immune cell populations. RESULTS [18F]AlF-mNOTA-GZP showed good predictive ability and correlated well with changes in tumour-associated T cells, especially CD8+ T cells; however, overall uptake and response to monotherapy or combination therapies was very different in the CT26 and MC38 tumours, likely due to the immunostimulatory environment imbued by the MSI-high phenotype in MC38 tumours. CONCLUSIONS [18F]AlF-mNOTA-GZP uptake correlates well with changes in CD8+ T cell populations and is able to stratify tumour response to a range of ICIs administered as monotherapies or in combination. However, tracer uptake can be significantly affected by preexisting phenotypic abnormalities potentially confusing data interpretation.
Collapse
Affiliation(s)
- J L Goggi
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - Y X Tan
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - S V Hartimath
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - B Jieu
- Institute of Chemical and Engineering Sciences (ICES), A*STAR, 8 Biomedical Grove, #07, Neuros, 138665, Singapore
| | - Y Y Hwang
- Singapore Immunology Network, A*STAR, 8A Biomedical Grove, Immunos, 138648, Singapore
| | - L Jiang
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - R Boominathan
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - P Cheng
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - T Y Yuen
- Institute of Chemical and Engineering Sciences (ICES), A*STAR, 8 Biomedical Grove, #07, Neuros, 138665, Singapore
| | - H X Chin
- Singapore Immunology Network, A*STAR, 8A Biomedical Grove, Immunos, 138648, Singapore
| | - J R Tang
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore
| | - A Larbi
- Singapore Immunology Network, A*STAR, 8A Biomedical Grove, Immunos, 138648, Singapore
| | - A M Chacko
- Laboratory for Translational and Molecular Imaging (LTMI), Cancer and Stem Cell Biology Programme, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - L Renia
- Singapore Immunology Network, A*STAR, 8A Biomedical Grove, Immunos, 138648, Singapore
| | - C Johannes
- p53 Laboratory, A*STAR, 8A Biomedical Grove, #06-04/05, Neuros/Immunos, 138665, Singapore
| | - Edward G Robins
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research (A* STAR), 11 Biopolis Way, #01-02, Helios, 138667, Singapore.
- Clinical Imaging Research Centre (CIRC), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
62
|
Gomez S, Tabernacki T, Kobyra J, Roberts P, Chiappinelli KB. Combining epigenetic and immune therapy to overcome cancer resistance. Semin Cancer Biol 2020; 65:99-113. [PMID: 31877341 PMCID: PMC7308208 DOI: 10.1016/j.semcancer.2019.12.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/02/2019] [Accepted: 12/19/2019] [Indexed: 01/09/2023]
Abstract
Cancer undergoes "immune editing" to evade destruction by cells of the host immune system including natural killer (NK) cells and cytotoxic T lymphocytes (CTLs). Current adoptive cellular immune therapies include CAR T cells and dendritic cell vaccines, strategies that have yet to show success for a wide range of tumors. Cancer resistance to immune therapy is driven by extrinsic factors and tumor cell intrinsic factors that contribute to immune evasion. These extrinsic factors include immunosuppressive cell populations such as regulatory T cells (Tregs), tumor-associated macrophages (TAMS), and myeloid-derived suppressor cells (MDSCs). These cells produce and secrete immunosuppressive factors and express inhibitory ligands that interact with receptors on T cells including PD-1 and CTLA-4. Immune checkpoint blockade (ICB) therapies such as anti-PD-1 and anti-CTLA-4 have shown success by increasing immune activation to eradicate cancer, though both primary and acquired resistance remain a problem. Tumor cell intrinsic factors driving primary and acquired resistance to these immune therapies include genetic and epigenetic mechanisms. Epigenetic therapies for cancer including DNA methyltransferase inhibitors (DNMTi), histone deacetylase inhibitors (HDACi), and histone methyltransferase inhibitors (HMTi) can stimulate anti-tumor immunity in both tumor cells and host immune cells. Here we discuss in detail tumor mechanisms of immune evasion and how common epigenetic therapies for cancer may be used to reverse immune evasion. Lastly, we summarize current clinical trials combining epigenetic therapies with immune therapies to reverse cancer immune resistance mechanisms.
Collapse
Affiliation(s)
- Stephanie Gomez
- The George Washington University Cancer Center, United States; The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, United States
| | - Tomasz Tabernacki
- The George Washington University Cancer Center, United States; The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, United States
| | - Julie Kobyra
- The George Washington University Cancer Center, United States; The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, United States
| | - Paige Roberts
- The George Washington University Cancer Center, United States; The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, United States
| | - Katherine B Chiappinelli
- The George Washington University Cancer Center, United States; The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, United States.
| |
Collapse
|
63
|
Campos Carrascosa L, van Beek AA, de Ruiter V, Doukas M, Wei J, Fisher TS, Ching K, Yang W, van Loon K, Boor PPC, Rakké YS, Noordam L, Doornebosch P, Grünhagen D, Verhoef K, Polak WG, IJzermans JNM, Ni I, Yeung YA, Salek-Ardakani S, Sprengers D, Kwekkeboom J. FcγRIIB engagement drives agonistic activity of Fc-engineered αOX40 antibody to stimulate human tumor-infiltrating T cells. J Immunother Cancer 2020; 8:jitc-2020-000816. [PMID: 32900860 PMCID: PMC7478034 DOI: 10.1136/jitc-2020-000816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2020] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND OX40 (CD134) is a costimulatory molecule of the tumor necrosis factor receptor superfamily that is currently being investigated as a target for cancer immunotherapy. However, despite promising results in murine tumor models, the clinical efficacy of agonistic αOX40 antibodies in the treatment of patients with cancer has fallen short of the high expectation in earlier-stage trials. METHODS Using lymphocytes from resected tumor, tumor-free (TF) tissue and peripheral blood mononuclear cells (PBMC) of 96 patients with hepatocellular and colorectal cancers, we determined OX40 expression and the in vitro T-cell agonistic activity of OX40-targeting compounds. RNA-Seq was used to evaluate OX40-mediated transcriptional changes in CD4+ and CD8+ human tumor-infiltrating lymphocytes (TILs). RESULTS Here, we show that OX40 was overexpressed on tumor-infiltrating CD4+ T cells compared with blood and TF tissue-derived T cells. In contrast to a clinical candidate αOX40 antibody, treatment with an Fc-engineered αOX40 antibody (αOX40_v12) with selectively enhanced FcγRIIB affinity, stimulated in vitro CD4+ and CD8+ TIL expansion, as well as cytokine and chemokine secretions. The activity of αOX40_v12 was dependent on FcγRIIB engagement and intrinsic CD3/CD28 signals. The transcriptional landscape of CD4+ and CD8+ TILs shifted toward a prosurvival, inflammatory and chemotactic profile on treatment with αOX40_v12. CONCLUSIONS OX40 is overexpressed on CD4+ TILs and thus represents a promising target for immunotherapy. Targeting OX40 with currently used agonistic antibodies may be inefficient due to lack of OX40 multimerization. Thus, Fc engineering is a powerful tool in enhancing the agonistic activity of αOX40 antibody and may shape the future design of antibody-mediated αOX40 immunotherapy.
Collapse
Affiliation(s)
- Lucia Campos Carrascosa
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Adriaan A van Beek
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Valeska de Ruiter
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Michail Doukas
- Pathology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Jie Wei
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | - Timothy S Fisher
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | - Keith Ching
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | - Wenjing Yang
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | - Karlijn van Loon
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Patrick P C Boor
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Yannick S Rakké
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Lisanne Noordam
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | | | - Dirk Grünhagen
- Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Kees Verhoef
- Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Wojciech G Polak
- Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Jan N M IJzermans
- Surgery, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Irene Ni
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | - Yik Andy Yeung
- Pfizer Cancer Immunology Discovery, Pfizer Inc, San Diego, California, USA
| | | | - Dave Sprengers
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Jaap Kwekkeboom
- Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
64
|
Haibe Y, El Husseini Z, El Sayed R, Shamseddine A. Resisting Resistance to Immune Checkpoint Therapy: A Systematic Review. Int J Mol Sci 2020; 21:E6176. [PMID: 32867025 PMCID: PMC7504220 DOI: 10.3390/ijms21176176] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 12/14/2022] Open
Abstract
The treatment landscape in oncology has witnessed a major revolution with the introduction of checkpoint inhibitors: anti-PD1, anti-PDL1 and anti-CTLA-4. These agents enhance the immune response towards cancer cells instead of targeting the tumor itself, contrary to standard chemotherapy. Although long-lasting durable responses have been observed with immune checkpoints inhibitors, the response rate remains relatively low in many cases. Some patients respond in the beginning but then eventually develop acquired resistance to treatment and progress. Other patients having primary resistance never respond. Multiple studies have been conducted to further elucidate these variations in response in different tumor types and different individuals. This paper provides an overview of the mechanisms of resistance to immune checkpoint inhibitors and highlights the possible therapeutic approaches under investigation aiming to overcome such resistance in order to improve the clinical outcomes of cancer patients.
Collapse
Affiliation(s)
| | | | | | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut-Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (Z.E.H.); (R.E.S.)
| |
Collapse
|
65
|
Zhuo M, Chi Y, Wang Z. The adverse events associated with combination immunotherapy in cancers: Challenges and chances. Asia Pac J Clin Oncol 2020; 16:e154-e159. [PMID: 32786161 DOI: 10.1111/ajco.13365] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/20/2020] [Indexed: 12/26/2022]
Abstract
With the development of cancer immunotherapy, the combination strategy is becoming prevalent. Multiple relevant clinical trials are ongoing in this field. However, immune-related adverse events (irAEs) occurred more frequently, showing a different pattern from single-agent therapy. It is necessary for clinicians to learn about the characteristics of AEs from combination immunotherapy, and master the skills to deal with them. In this article, we reviewed presently published data about AEs from combination immunotherapy of cancers. We believe a full-scale view about this new treatment strategy will facilitate oncologists to better understand tumor immune response. With cutting edge knowledge, an experienced team can minimize these AEs and help patients to achieve high-quality long-term survival.
Collapse
Affiliation(s)
- Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yujia Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Ziping Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|
66
|
Rocco D, Gregorc V, Della Gravara L, Lazzari C, Palazzolo G, Gridelli C. New immunotherapeutic drugs in advanced non-small cell lung cancer (NSCLC): from preclinical to phase I clinical trials. Expert Opin Investig Drugs 2020; 29:1005-1023. [PMID: 32643447 DOI: 10.1080/13543784.2020.1793956] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The development of immune checkpoint inhibitors (ICI) has represented a revolution in the treatment of non-small cell lung cancer (NSCLC) and has established a new standard of care for different settings. However, through adaptive changes, cancer cells can develop resistance mechanisms to these drugs, hence the necessity for novel immunotherapeutic agents. AREAS COVERED This paper explores the immunotherapeutics currently under investigation in phase I clinical trials for the treatment of NSCLC as monotherapies and combination therapies. It provides two comprehensive tables of phase I agents which are listed according to target, drug, drug class, mechanism of action, setting, trial identifier, and trial status. A comprehensive literature search was carried out to identify eligible studies from MEDLINE/PubMed and ClinicalTrials.gov. EXPERT OPINION A key hurdle to success in this field is our limited understanding of the synergic interactions of the immune targets in the context of the TME. While we can recognize the links between inhibitors and some particularly promising new targets such as TIM-3 and LAG3, we continue to develop approaches to exploit their interactions to enhance the immune response of the patient to tumor cells.
Collapse
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN Dei Colli Monaldi , Naples, Italy
| | - Vanesa Gregorc
- Department of Oncology, Division of Experimental Medicine, IRCCS San Raffaele , Milan, Italy
| | - Luigi Della Gravara
- Department of Experimental Medicine, Università Degli Studi Della Campania "Luigi Vanvitelli" , Caserta, Italy
| | - Chiara Lazzari
- Department of Oncology, Division of Experimental Medicine, IRCCS San Raffaele , Milan, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, "S.G. Moscati" Hospital , Avellino, Italy
| |
Collapse
|
67
|
Advances in Anti-Cancer Immunotherapy: Car-T Cell, Checkpoint Inhibitors, Dendritic Cell Vaccines, and Oncolytic Viruses, and Emerging Cellular and Molecular Targets. Cancers (Basel) 2020; 12:cancers12071826. [PMID: 32645977 PMCID: PMC7408985 DOI: 10.3390/cancers12071826] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Unlike traditional cancer therapies, such as surgery, radiation and chemotherapy that are typically non-specific, cancer immunotherapy harnesses the high specificity of a patient’s own immune system to selectively kill cancer cells. The immune system is the body’s main cancer surveillance system, but cancers may evade destruction thanks to various immune-suppressing mechanisms. We therefore need to deploy various immunotherapy-based strategies to help bolster the anti-tumour immune responses. These include engineering T cells to express chimeric antigen receptors (CARs) to specifically recognise tumour neoantigens, inactivating immune checkpoints, oncolytic viruses and dendritic cell (DC) vaccines, which have all shown clinical benefit in certain cancers. However, treatment efficacy remains poor due to drug-induced adverse events and immunosuppressive tendencies of the tumour microenvironment. Recent preclinical studies have unveiled novel therapies such as anti-cathepsin antibodies, galectin-1 blockade and anti-OX40 agonistic antibodies, which may be utilised as adjuvant therapies to modulate the tumour microenvironment and permit more ferocious anti-tumour immune response.
Collapse
|
68
|
Poropatich K, Dominguez D, Chan WC, Andrade J, Zha Y, Wray B, Miska J, Qin L, Cole L, Coates S, Patel U, Samant S, Zhang B. OX40+ plasmacytoid dendritic cells in the tumor microenvironment promote antitumor immunity. J Clin Invest 2020; 130:3528-3542. [PMID: 32182225 PMCID: PMC7324178 DOI: 10.1172/jci131992] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 03/11/2020] [Indexed: 12/11/2022] Open
Abstract
Plasmacytoid DCs (pDCs), the major producers of type I interferon, are principally recognized as key mediators of antiviral immunity. However, their role in tumor immunity is less clear. Depending on the context, pDCs can promote or suppress antitumor immune responses. In this study, we identified a naturally occurring pDC subset expressing high levels of OX40 (OX40+ pDC) enriched in the tumor microenvironment (TME) of head and neck squamous cell carcinoma. OX40+ pDCs were distinguished by a distinct immunostimulatory phenotype, cytolytic function, and ability to synergize with conventional DCs (cDCs) in generating potent tumor antigen-specific CD8+ T cell responses. Transcriptomically, we found that they selectively utilized EIF2 signaling and oxidative phosphorylation pathways. Moreover, depletion of pDCs in the murine OX40+ pDC-rich tumor model accelerated tumor growth. Collectively, we present evidence of a pDC subset in the TME that favors antitumor immunity.
Collapse
Affiliation(s)
- Kate Poropatich
- Department of Pathology
- Robert H. Lurie Comprehensive Cancer Center, and
| | - Donye Dominguez
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | | | | | - Yuanyuan Zha
- Human Immunologic Monitoring Facility, Office of Shared Research Facilities, University of Chicago, Chicago, Illinois, USA
| | - Brian Wray
- Department of Biochemistry and Molecular Genetics
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lei Qin
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Lisa Cole
- Department of Chemistry, Northwestern University, Evanston, Illinois, USA
| | - Sydney Coates
- Head and Neck Surgery, Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Urjeet Patel
- Robert H. Lurie Comprehensive Cancer Center, and
- Head and Neck Surgery, Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sandeep Samant
- Robert H. Lurie Comprehensive Cancer Center, and
- Head and Neck Surgery, Department of Otolaryngology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Bin Zhang
- Robert H. Lurie Comprehensive Cancer Center, and
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
69
|
Jordan AC, Wu J. Immunotherapy in hepatocellular carcinoma: Combination strategies. World J Meta-Anal 2020; 8:190-209. [DOI: 10.13105/wjma.v8.i3.190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
Liver cancer is one of the most common causes of cancer death globally, and its incidence in the United States is increasing. Patients with advanced hepatocellular carcinoma (HCC) who are not candidates for surgical resection, liver transplant, or locoregional therapies can be treated with systemic therapies. Multiple agents, including sorafenib, lenvatinib, and regorafenib are approved for use as either first- or second-line therapy in this patient population, but all have relatively modest survival benefits. HCC is potentially susceptible to therapy with checkpoint inhibitors, including agents such as nivolumab and pembrolizumab, which are both approved by the Food and Drug Administration for patients previously treated with sorafenib but have not demonstrated superior overall survival in phase III trials. It is clear that more effective approaches are needed to potentiate the effects of checkpoint inhibitors in patients with HCC. This review will outline and appraise the current literature on the use of checkpoint inhibitors in HCC as part of a combination treatment involving an additional mode of therapy. The list of agents that can be paired with checkpoint inhibitors includes an additional checkpoint inhibitor, vascular endothelial growth factor or vascular endothelial growth factor receptor inhibitors, tyrosine kinase inhibitors, OX-40 agonists, and PT-112 inhibitors. The main non-pharmacologic therapies currently being studied for inclusion in a combination strategy include radiation therapy, trans-arterial chemoembolization, and ablation.
Collapse
Affiliation(s)
- Alexander Claudius Jordan
- Department of Internal Medicine, New York University School of Medicine, New York, NY 10016, United States
| | - Jennifer Wu
- Division of Hematology and Oncology, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, United States
| |
Collapse
|
70
|
Immune checkpoint inhibition in myeloid malignancies: Moving beyond the PD-1/PD-L1 and CTLA-4 pathways. Blood Rev 2020; 45:100709. [PMID: 32487480 DOI: 10.1016/j.blre.2020.100709] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/26/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICI) have yielded mixed but largely underwhelming results in clinical trials in patients with acute myeloid leukemia and myelodysplastic syndromes to date. However, increasing understanding of the immunologic landscape, potential biomarkers for benefits, and mechanisms of resistance, as well as the use of rational combinations, and identification of novel targets leaves plenty of room for optimism. Herein, we review recent advances in the preclinical and clinical development of ICI therapy in patients with myeloid malignancies and explore some of the important challenges facing the field such as the absence of validated biomarkers, the development of synergistic and safe combination therapies, and efforts to determine the best setting of ICI along the disease course. We finally foresee the future of the field and propose solutions to some of the major beforementioned obstacles.
Collapse
|
71
|
Gu S, Zi J, Han Q, Song C, Ge Z. Elevated TNFRSF4 gene expression is a predictor of poor prognosis in non-M3 acute myeloid leukemia. Cancer Cell Int 2020; 20:146. [PMID: 32390761 PMCID: PMC7197135 DOI: 10.1186/s12935-020-01213-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/15/2020] [Indexed: 01/15/2023] Open
Abstract
Background We used bioinformatic tools to dichotomize 157 non-M3 AML patients from the TCGA dataset based on the presence or absence of TP53 mutations, and screened out a key gene related to TP53 mutation for future analysis. Methods DEGs were analyzed by R package “DESeq2” and then run GSEA, GO enrichment, KEGG pathway and PPI network. Hub genes were selected out according to MCC. Log-rank (Mantel–Cox) test was used for survival analysis. Mann–Whitney U’s nonparametric t test and Fisher’s exact test was used for continuous and categorical variables respectively. p value< 0.05 was considered to be statistical significance. Results TNFRSF4 was final screened out as a key gene. Besides TP53 mutation (p = 0.0118), high TNFRSF4 was also associated with FLT3 mutation (p = 0.0102) and NPM1 mutation (p = 0.0024). Elevated TNFRSF4 was significantly related with intermediate (p = 0.0004) and poor (p = 0.0011) risk stratification as well as relapse statute (p = 0.0099). Patients with elevated TNFRSF4 expression had significantly shorter overall survival (median survival: 2.35 months vs. 21 months, p < 0.0001). Based on our clinical center data, TNFRSF4 expression was significantly higher in non-M3 AML patients than HDs (p = 0.0377) and MDS patients (EB-1, 2; p = 0.0017). Conclusions Elevated TNFRSF4 expression was associated with TP53, FLT3 and NPM1 mutation as well as poor clinical outcome. TNFRSF4 expression was significantly higher in non-M3 AML patients than HDs and MDS (EB-1, 2) patients. TNFRSF4 is need for future functional and mechanistic studies to investigate the role in non-M3 AML.
Collapse
Affiliation(s)
- Siyu Gu
- 1Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, No. 87, Dingjiaqiao, Gulou District, Nanjing, 210009 Jiangsu China
| | - Jie Zi
- 1Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, No. 87, Dingjiaqiao, Gulou District, Nanjing, 210009 Jiangsu China
| | - Qi Han
- 1Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, No. 87, Dingjiaqiao, Gulou District, Nanjing, 210009 Jiangsu China
| | - Chunhua Song
- 2Hershey Medical Center, Pennsylvania State University Medical College, Hershey, PA17033 USA
| | - Zheng Ge
- 1Department of Hematology, Zhongda Hospital, School of Medicine, Southeast University, Institute of Hematology Southeast University, No. 87, Dingjiaqiao, Gulou District, Nanjing, 210009 Jiangsu China
| |
Collapse
|
72
|
Zang H, Peng J, Zheng H, Fan S. Hyperprogression After Immune-Checkpoint Inhibitor Treatment: Characteristics and Hypotheses. Front Oncol 2020; 10:515. [PMID: 32411591 PMCID: PMC7201048 DOI: 10.3389/fonc.2020.00515] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 03/23/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapies in tumors have attracted increasing attention. They play an important role in precision medicine. Many immune-checkpoint inhibitors (ICIs) have obtained FDA approval and show good performance in the clinic. Hyperprogressive disease (HPD) after ICIs was first described in November 2016. Since then, a series of cases of HPD after ICIs have been reported. Notwithstanding that only a small subset of patients may experience this atypical response, HPD in affected patients means shorter survival times and worse prognoses. We summarized common standards for HPD diagnosis and profiled advantages and disadvantages. Elderly age, MDM2 family amplification, infiltration of PD-1-positive regulatory effector T cells and M2-like macrophages, and cancer stem cells may take part in HPD occurrence. Overall, we should focus on investigating the early markers and pathogenic mechanisms of HPD to solve this issue in ICIs.
Collapse
Affiliation(s)
- Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Basic Medical School, Central South University, Changsha, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
73
|
Mahmood J, Alexander AA, Samanta S, Kamlapurkar S, Singh P, Saeed A, Carrier F, Cao X, Shukla HD, Vujaskovic Z. A Combination of Radiotherapy, Hyperthermia, and Immunotherapy Inhibits Pancreatic Tumor Growth and Prolongs the Survival of Mice. Cancers (Basel) 2020; 12:cancers12041015. [PMID: 32326142 PMCID: PMC7226594 DOI: 10.3390/cancers12041015] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/03/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Pancreatic cancer (PC) is the fourth-most-deadly cancer in the United States with a 5-year survival rate of only 8%. Unfortunately, only 10–20% of PC patients are candidates for surgery, with the vast majority of patients with locally-advanced disease undergoing chemotherapy and/or radiation therapy (RT). Current treatments are clearly inadequate and novel strategies are crucially required. We investigated a novel tripartite treatment (combination of tumor targeted hyperthermia (HT), radiation therapy (RT), and immunotherapy (IT)) to alter immunosuppressive PC-tumor microenvironment (TME). (2). Methods: In a syngeneic PC murine tumor model, HT was delivered before tumor-targeted RT, by a small animal radiation research platform (SARRP) followed by intraperitoneal injections of cytotoxic T-cell agonist antibody against OX40 (also known as CD134 or Tumor necrosis factor receptor superfamily member 4; TNFRSF4) that can promote T-effector cell activation and inhibit T-regulatory (T-reg) function. (3). Results: Tripartite treatment demonstrated significant inhibition of tumor growth (p < 0.01) up to 45 days post-treatment with an increased survival rate compared to any monotherapy. Flow cytometric analysis showed a significant increase (p < 0.01) in cytotoxic CD8 and CD4+ T-cells in the TME of the tripartite treatment groups. There was no tripartite-treatment-related toxicity observed in mice. (4). Conclusions: Tripartite treatment could be a novel therapeutic option for PC patients.
Collapse
Affiliation(s)
- Javed Mahmood
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
- Correspondence: ; Tel.: +1-410-706-5133
| | - Allen A. Alexander
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - Santanu Samanta
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - Shriya Kamlapurkar
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - Prerna Singh
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - Ali Saeed
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - France Carrier
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| | - Xuefang Cao
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Hem D Shukla
- Department of Neurology and Neurosurgery, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA;
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences (DTRS), Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.A.A.); (S.S.); (S.K.); (P.S.); (A.S.); (F.C.); (Z.V.)
| |
Collapse
|
74
|
Han Y, Zhu L, Wu W, Zhang H, Hu W, Dai L, Yang Y. Small Molecular Immune Modulators as Anticancer Agents. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:547-618. [PMID: 32185725 DOI: 10.1007/978-981-15-3266-5_22] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
After decades of intense effort, immune checkpoint inhibitors have been conclusively demonstrated to be effective in cancer treatments and thus are revolutionizing the concepts in the treatment of cancers. Immuno-oncology has arrived and will play a key role in cancer treatment in the foreseeable future. However, efforts to find novel methods to improve the immune response to cancer have not ceased. Small-molecule approaches offer inherent advantages over biologic immunotherapies since they can cross cell membranes, penetrate into tumor tissue and tumor microenvironment more easily, and are amenable to be finely controlled than biological agents, which may help reduce immune-related adverse events seen with biologic therapies and provide more flexibility for the combination use with other therapies and superior clinical benefit. On the one hand, small-molecule therapies can modulate the immune response to cancer by restoring the antitumor immunity, promoting more effective cytotoxic lymphocyte responses, and regulating tumor microenvironment, either directly or epigenetically. On the other hand, the combination of different mechanisms of small molecules with antibodies and other biologics demonstrated admirable synergistic effect in clinical settings for cancer treatment and may expand antibodies' usefulness for broader clinical applications. This chapter provides an overview of small-molecule immunotherapeutic approaches either as monotherapy or in combination for the treatment of cancer.
Collapse
Affiliation(s)
- Yongxin Han
- Lapam Capital LLC., 17C1, Tower 2, Xizhimenwai Street, Xicheng District, Beijing, 100044, China.
| | - Li Zhu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Wu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Hui Zhang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Wei Hu
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Liguang Dai
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| | - Yanqing Yang
- PrimeGene (Beijing) Co., Ltd., Fengtai District, Beijing, 100070, China
| |
Collapse
|
75
|
Discovery of New Immune Checkpoints: Family Grows Up. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1248:61-82. [PMID: 32185707 DOI: 10.1007/978-981-15-3266-5_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The first generation of immune checkpoint inhibitors (ICIs) including anti-CTLA-4 and anti-PD-1/anti-PD-L1 has achieved profound and great success. Till 2019 Q1, there are nine ICIs landing the oncology market: Ipilimumab (anti-CTLA-4, Bristol-Myers Squibb), Nivolumab (anti-PD-1, Bristol-Myers Squibb), Pembrolizumab (anti-PD-1, Merck), Atezolizumab (anti-PD-L1, Roche/Genentech), Durvalumab (anti-PD-L1, Astra Zeneca), Tremelimumab (anti-CTLA-4, Astra Zeneca), Cemiplimab (anti-PD-1, Sanofi/Regeneron), Toripalimab (anti-PD-1, Junshi), and Sintilimab (anti-PD-1, Innovent), which have covered the majority of hematologic and solid malignancies' indication. Beyond the considerable benefits for the patients, frustrated boundary still exists: limited response rate in monotherapy in late-stage population, poor effectiveness in neoplasms with immune desert and immune excluded types, and immune-related toxicities, some are life-threatened and with higher incidence in I-O combination regiment. Moreover, clinicians observed some cases switching to progression after achieving partial or complete response, indicating treatment failure or drug resistance. So people begin looking for the next generation of immune checkpoint members.
Collapse
|
76
|
Gaspar M, Pravin J, Rodrigues L, Uhlenbroich S, Everett KL, Wollerton F, Morrow M, Tuna M, Brewis N. CD137/OX40 Bispecific Antibody Induces Potent Antitumor Activity that Is Dependent on Target Coengagement. Cancer Immunol Res 2020; 8:781-793. [PMID: 32273279 DOI: 10.1158/2326-6066.cir-19-0798] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/31/2020] [Accepted: 03/31/2020] [Indexed: 11/16/2022]
Abstract
Following the success of immune checkpoint blockade therapy against cancer, agonistic antibodies targeting T-cell costimulatory pathways are in clinical trials. The TNF superfamily of receptors (TNFRSF) members CD137 and OX40 are costimulatory receptors that stimulate T-cell proliferation and activation upon interaction with their cognate ligands. Activating CD137 and OX40 with agonistic mAbs stimulates the immune system due to their broad expression on CD4+ and CD8+ T cells and natural killer cells and has antitumor effects in preclinical models. Most TNFRSF agonist antibodies require crosslinking via Fcγ receptors (FcγR), which can limit their clinical activity. FS120 mAb2, a dual agonist bispecific antibody targeting CD137 and OX40, activated both CD4+ and CD8+ T cells in an FcγR-independent mechanism, dependent on concurrent binding. A mouse surrogate version of the bispecific antibody displayed antitumor activity in syngeneic tumor models, independent of T regulatory cell depletion and of FcγR interaction, but associated with peripheral T-cell activation and proliferation. When compared with a crosslink-independent CD137 agonist mAb, the FS120 surrogate induced lower liver T-cell infiltration. These data support initiation of clinical development of FS120, a first-in-class dual agonist bispecific antibody for the treatment of human cancer.
Collapse
Affiliation(s)
| | - John Pravin
- F-star Therapeutics Ltd., Cambridge, United Kingdom
| | | | | | | | | | | | | | - Neil Brewis
- F-star Therapeutics Ltd., Cambridge, United Kingdom.
| |
Collapse
|
77
|
Coleman MF, Cozzo AJ, Pfeil AJ, Etigunta SK, Hursting SD. Cell Intrinsic and Systemic Metabolism in Tumor Immunity and Immunotherapy. Cancers (Basel) 2020; 12:cancers12040852. [PMID: 32244756 PMCID: PMC7225951 DOI: 10.3390/cancers12040852] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has shown extraordinary promise at treating cancers otherwise resistant to treatment. However, for ICI therapy to be effective, it must overcome the metabolic limitations of the tumor microenvironment. Tumor metabolism has long been understood to be highly dysregulated, with potent immunosuppressive effects. Moreover, T cell activation and longevity within the tumor microenvironment are intimately tied to T cell metabolism and are required for the long-term efficacy of ICI therapy. We discuss in this review the intersection of metabolic competition in the tumor microenvironment, T cell activation and metabolism, the roles of tumor cell metabolism in immune evasion, and the impact of host metabolism in determining immune surveillance and ICI therapy outcomes. We also discussed the effects of obesity and calorie restriction—two important systemic metabolic perturbations that impact intrinsic metabolic pathways in T cells as well as cancer cells.
Collapse
Affiliation(s)
- Michael F. Coleman
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Alyssa J. Cozzo
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Department of Medicine, Duke University, Durham, NC 27705, USA
| | - Alexander J. Pfeil
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Suhas K. Etigunta
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC 27516, USA; (M.F.C.); (A.J.C.); (A.J.P.); (S.K.E.)
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27516, USA
- Correspondence:
| |
Collapse
|
78
|
Fu Y, Lin Q, Zhang Z, Zhang L. Therapeutic strategies for the costimulatory molecule OX40 in T-cell-mediated immunity. Acta Pharm Sin B 2020; 10:414-433. [PMID: 32140389 PMCID: PMC7049610 DOI: 10.1016/j.apsb.2019.08.010] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
The T cell co-stimulatory molecule OX40 and its cognate ligand OX40L have attracted broad research interest as a therapeutic target in T cell-mediated diseases. Accumulating preclinical evidence highlights the therapeutic efficacy of both agonist and blockade of the OX40-OX40L interaction. Despite this progress, many questions about the immuno-modulator roles of OX40 on T cell function remain unanswered. In this review we summarize the impact of the OX40-OX40L interaction on T cell subsets, including Th1, Th2, Th9, Th17, Th22, Treg, Tfh, and CD8+ T cells, to gain a comprehensive understanding of anti-OX40 mAb-based therapies. The potential therapeutic application of the OX40-OX40L interaction in autoimmunity diseases and cancer immunotherapy are further discussed; OX40-OX40L blockade may ameliorate autoantigen-specific T cell responses and reduce immune activity in autoimmunity diseases. We also explore the rationale of targeting OX40-OX40L interactions in cancer immunotherapy. Ligation of OX40 with targeted agonist anti-OX40 mAbs conveys activating signals to T cells. When combined with other therapeutic treatments, such as anti-PD-1 or anti-CTLA-4 blockade, cytokines, chemotherapy, or radiotherapy, the anti-tumor activity of agonist anti-OX40 treatment will be further enhanced. These data collectively suggest great potential for OX40-mediated therapies.
Collapse
Affiliation(s)
- Yu Fu
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Qing Lin
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Ling Zhang
- Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, College of Polymer Science and Engineering, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| |
Collapse
|
79
|
Quintella CM, Quintella HM, Rohweder M, Quintella GM. Advances in patent applications related to cancer vaccine using CpG-ODN and OX40 association. Expert Opin Ther Pat 2020; 30:287-301. [PMID: 32008403 DOI: 10.1080/13543776.2020.1724960] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: This review aims to assess the available technologies, advances, and trends from technological readiness level 4 to level 8 for cancer immunologic therapeutics using the association of OX40 and CPG-ODN, usually known as cancer vaccine.Areas covered: Patent documents and clinic studies referring to the use of CpG-ODN and of OX40 association for cancer therapeutics. Patent data were obtained within the worldwide basis of the European Patent Office (EPO). The 138 patents of 36 patent families found were analyzed focusing on word distribution of technology developers and potential markets, legal status, annual evolution of first priority, technological domains, applicants and co-applicants and detailed analysis of each technology. Two clinical studies are in progress.Expert opinion: Traditional methods in post cancer diagnosis are being replaced by immunological association therapies. It is expected that the development of cancer vaccines will expand the scope of cancer-specific immunotherapy, especially if associated with alternative systems for expression and delivery with future potential. It is expected that genetic and controlled and/or specific nano delivery are improved. Furthermore, these new developments will likely address the problem of long-term treatments, reducing cancer mortality and reducing patient numbers worldwide.
Collapse
Affiliation(s)
- Cristina M Quintella
- Chemistry Institute, Federal University of Bahia, Campus Universitário de Ondina, Salvador, BA, Brasil.,Medicine School, Federal University of Minas Gerais, Belo Horizonte, MG, Brasil
| | - Heitor M Quintella
- PROFNIT - Postgraduate Program on Intellectual Property and Technology Transfer for Innovation, Federal University of Bahia, Campus Universitário de Ondina, Salvador, BA, Brasil
| | - Mayla Rohweder
- Chemistry Institute, Federal University of Bahia, Campus Universitário de Ondina, Salvador, BA, Brasil.,Medicine School, Federal University of Minas Gerais, Belo Horizonte, MG, Brasil.,CEPARH - Research and Assistance Center on Human Reproduction, Salvador, BA, Brazil
| | - Guilherme M Quintella
- Chemistry Institute, Federal University of Bahia, Campus Universitário de Ondina, Salvador, BA, Brasil.,Medicine School, Federal University of Minas Gerais, Belo Horizonte, MG, Brasil.,Quintellar Legal Consulting Company, Salvador, BA, Brazil
| |
Collapse
|
80
|
Jeong S, Park SH. Co-Stimulatory Receptors in Cancers and Their Implications for Cancer Immunotherapy. Immune Netw 2020; 20:e3. [PMID: 32158591 PMCID: PMC7049585 DOI: 10.4110/in.2020.20.e3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 01/27/2020] [Accepted: 01/27/2020] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs), including anti-PD-1 and anti-CTLA-4 therapeutic agents, are now approved by the Food and Drug Administration for treatment of various types of cancer. However, the therapeutic efficacy of ICIs varies among patients and cancer types. Moreover, most patients do not develop durable antitumor responses after ICI therapy due to an ephemeral reversal of T-cell dysfunction. As co-stimulatory receptors play key roles in regulating the effector functions of T cells, activating co-stimulatory pathways may improve checkpoint inhibition efficacy, and lead to durable antitumor responses. Here, we review recent advances in our understating of co-stimulatory receptors in cancers, providing the necessary groundwork for the rational design of cancer immunotherapy.
Collapse
Affiliation(s)
- Seongju Jeong
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea
| | - Su-Hyung Park
- Biomedical Science and Engineering Interdisciplinary Program, KAIST, Daejeon 34141, Korea.,Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| |
Collapse
|
81
|
Canel M, Taggart D, Sims AH, Lonergan DW, Waizenegger IC, Serrels A. T-cell co-stimulation in combination with targeting FAK drives enhanced anti-tumor immunity. eLife 2020; 9:e48092. [PMID: 31959281 PMCID: PMC6974352 DOI: 10.7554/elife.48092] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023] Open
Abstract
Focal Adhesion Kinase (FAK) inhibitors are currently undergoing clinical testing in combination with anti-PD-1 immune checkpoint inhibitors. However, which patients are most likely to benefit from FAK inhibitors, and what the optimal FAK/immunotherapy combinations are, is currently unknown. We identify that cancer cell expression of the T-cell co-stimulatory ligand CD80 sensitizes murine tumors to a FAK inhibitor and show that CD80 is expressed by human cancer cells originating from both solid epithelial cancers and some hematological malignancies in which FAK inhibitors have not been tested clinically. In the absence of CD80, we identify that targeting alternative T-cell co-stimulatory receptors, in particular OX-40 and 4-1BB in combination with FAK, can drive enhanced anti-tumor immunity and even complete regression of murine tumors. Our findings provide rationale supporting the clinical development of FAK inhibitors in combination with patient selection based on cancer cell CD80 expression, and alternatively with therapies targeting T-cell co-stimulatory pathways.
Collapse
Affiliation(s)
- Marta Canel
- Centre for Inflammation Research, Queen’s Medical Research InstituteUniversity of EdinburghEdinburghUnited Kingdom
| | - David Taggart
- Centre for Inflammation Research, Queen’s Medical Research InstituteUniversity of EdinburghEdinburghUnited Kingdom
| | - Andrew H Sims
- Cancer Research UK Edinburgh Centre, MRC Institute of Genetics & Molecular MedicineUniversity of EdinburghEdinburghUnited Kingdom
| | - David W Lonergan
- Centre for Inflammation Research, Queen’s Medical Research InstituteUniversity of EdinburghEdinburghUnited Kingdom
| | | | - Alan Serrels
- Centre for Inflammation Research, Queen’s Medical Research InstituteUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
82
|
Heinhuis KM, Carlino M, Joerger M, Di Nicola M, Meniawy T, Rottey S, Moreno V, Gazzah A, Delord JP, Paz-Ares L, Britschgi C, Schilder RJ, O'Byrne K, Curigliano G, Romano E, Patah P, Wang R, Liu Y, Bajaj G, Siu LL. Safety, Tolerability, and Potential Clinical Activity of a Glucocorticoid-Induced TNF Receptor-Related Protein Agonist Alone or in Combination With Nivolumab for Patients With Advanced Solid Tumors: A Phase 1/2a Dose-Escalation and Cohort-Expansion Clinical Trial. JAMA Oncol 2020; 6:100-107. [PMID: 31697308 DOI: 10.1001/jamaoncol.2019.3848] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance Multiple immunostimulatory agonist antibodies have been clinically tested in solid tumors to evaluate the role of targeting glucocorticoid-induced tumor necrosis factor (TNF) receptor-related protein in anticancer treatments. Objective To evaluate the safety and activity of the fully human glucocorticoid-induced TNF receptor-related protein agonist IgG1 monoclonal antibody BMS-986156 with or without nivolumab in patients with advanced solid tumors. Design, Setting, and Participants This global, open-label, phase 1/2a study of BMS-986156 with or without nivolumab enrolled 292 patients 18 years or older with advanced solid tumors and an Eastern Cooperative Oncology Group performance status of 1 or less. Prior checkpoint inhibitor therapy was allowed. Monotherapy and combination dose-escalation cohorts ran concurrently to guide expansion doses beginning October 16, 2015; the study is ongoing. Interventions The protein agonist BMS-986156 was administered intravenously at a dose of 10, 30, 100, 240, or 800 mg every 2 weeks as monotherapy, and in the combination group 30, 100, 240, or 800 mg plus 240 mg of nivolumab every 2 weeks; same-dose cohorts were pooled for analysis. One cohort also received 480 mg of BMS-986156 plus 480 mg of nivolumab every 4 weeks. Main Outcomes and Measures The primary end points were safety, tolerability, and dose-limiting toxic effects. Additional end points included antitumor activity per Response Evaluation Criteria in Solid Tumors, version 1.1, and exploratory biomarker analyses. Results With a follow-up range of 1.4 to 101.7 weeks (follow-up ongoing), 34 patients (16 women and 18 men; median age, 56.6 years [range, 28-75 years]) received monotherapy (4 patients completed initial treatment), and 258 patients (140 women and 118 men; median age, 60 years [range, 21-87 years]) received combination therapy (65 patients completed initial treatment). No grade 3 to 5 treatment-related adverse events occurred with BMS-986156 monotherapy; grade 3 to 4 treatment-related adverse events occurred in 24 patients (9.3%) receiving BMS-986156 plus nivolumab, with no grade 5 treatment-related adverse events. One dose-limiting toxic effect (grade 4 elevated creatine phosphokinase levels) occurred in a patient receiving 800 mg of BMS-986156 plus 240 mg of nivolumab every 2 weeks; BMS-986156 with or without nivolumab exhibited linear pharmacokinetics with dose-related increase after a single dose. Peripheral T-cell and natural killer-cell proliferation increased after administration of BMS-986156 with or without nivolumab. No consistent and significant modulation of intratumoral CD8+ T cells and FoxP3+ regulatory T cells was observed. No responses were seen with BMS-986156 alone; objective response rates ranged from 0% to 11.1% (1 of 9) across combination therapy cohorts, with a few responses observed in patients previously treated with anti-programmed death receptor (ligand) 1 therapy. Conclusions and Relevance Based on this cohort, BMS-986156 appears to have had a manageable safety profile, and BMS-986156 plus nivolumab demonstrated safety and efficacy comparable to historical data reported for nivolumab monotherapy. Trial Registration ClinicalTrials.gov identifier: NCT02598960.
Collapse
Affiliation(s)
- Kimberley M Heinhuis
- Division of Pharmacology, The Netherlands Cancer Institute Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Matteo Carlino
- Department of Medical Oncology, Crown Princess Mary Cancer Centre Westmead Hospital, Westmead, Australia
| | - Markus Joerger
- Department of Internal Medicine, Clinic for Medical Oncology and Hematology, Cantonal Hospital St Gallen, St Gallen, Switzerland
| | - Massimo Di Nicola
- Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale dei Tumori Milano, Milano, Italy
| | | | - Sylvie Rottey
- Department of Medical Oncology, Universitair Ziekenhuis Ghent, Ghent, Belgium
| | - Victor Moreno
- South Texas Accelerated Research Therapeutics Madrid-Fundacion Jimenez Diaz, Fundacion Jimenez Diaz Hospital, Madrid, Spain
| | - Anas Gazzah
- Drug Development Department, Gustave Roussy, Villejuif, France
| | - Jean-Pierre Delord
- Medical Oncology Departement, Institut Claudius Regaud and Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France
| | - Luis Paz-Ares
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Christian Britschgi
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Russell J Schilder
- Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - Kenneth O'Byrne
- Princess Alexandra Hospital and Queensland University of Technology, Brisbane, Australia
| | - Giuseppe Curigliano
- New Drugs Development Division for Innovative Therapies, University of Milano and Istituto Europeo Di Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Milano, Italy
| | - Emanuela Romano
- Department of Oncology, Center of Cancer Immunotherapy, U932, Institut Curie, Paris, France
| | | | - Rui Wang
- Bristol-Myers Squibb, Princeton, New Jersey
| | - Yali Liu
- Bristol-Myers Squibb, Princeton, New Jersey
| | | | - Lillian L Siu
- Bras and Family Drug Development Program, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
83
|
Overview of Basic Immunology and Clinical Application. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1244:1-36. [PMID: 32301008 DOI: 10.1007/978-3-030-41008-7_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor exists as a complex network of structures with an ability to evolve and evade the host immune surveillance mechanism. The immune milieu which includes macrophages, dendritic cells, natural killer cells, neutrophils, mast cells, B cells, and T cells are found in the core, the invasive margin, or the adjacent stromal or lymphoid component of the tumor. The immune infiltrate is heterogeneous and varies within a patient and between patients of the same tumor histology. The location, density, functionality, and cross-talk between the immune cells in the tumor microenvironment influence the nature of immune response, prognosis, and treatment outcomes in cancer patients. Therefore, an understanding of the characteristics of the immune cells and their role in tumor immune surveillance is of paramount importance to identify immune targets and to develop novel immune therapeutics in the war against cancer. In this chapter, we provide an overview of the individual components of the human immune system and the translational relevance of predictive biomarkers.
Collapse
|
84
|
High OX-40 expression in the tumor immune infiltrate is a favorable prognostic factor of overall survival in non-small cell lung cancer. J Immunother Cancer 2019; 7:351. [PMID: 31843013 PMCID: PMC6915970 DOI: 10.1186/s40425-019-0827-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction OX-40 co-stimulatory signaling plays a role in mounting anti-tumor immune responses and clinical trials targeting this pathway are ongoing. However, the association of with OX-40 protein expression with clinical outcomes and pathological features in non-small cell lung cancer (NSCLC) are largely unknown. Methods Surgically-resected stage I-III NSCLC specimens (N = 100) were stained by immunohistochemistry (IHC) for the following immune markers: OX-40, PD-L1, PD-1, CD3, CD4, CD8, CD45RO, CD57, CD68, FOXP3, granzyme B, and ICOS. Immune-related markers mRNA expression were also assessed. We evaluated the association of OX-40 levels with major clinicopathologic variables, including molecular driver mutations. Results OX-40 IHC expression was observed in all tested tumors, predominantly localized in the membrane of the tumor immune infiltrate, and was not associated with a specific clinicopathologic or molecular subtype. High OX-40 expression levels measured by IHC median score were associated with better overall survival (OS) (p = 0.002), independent of CD3/CD8, PD-L1, and ICOS expression. High OX-40 IHC score was associated with increased expression of immune-related genes such as CD3, IFN-gamma, ICOS, CD8, CXCL9, CXCL10, CCL5, granzyme K. Conclusions High OX-40 IHC expression in the tumor immune infiltrate is associated with favorable prognosis and increased levels of immune-related genes including IFN-gamma in patients with surgically resected stage I-III NSCLC. Its prognostic utility is independent of PD-L1 and other common markers of immune activation. High OX-40 expression potentially identifies a unique subgroup of NSCLC that may benefit from co-stimulation with OX-40 agonist antibodies and potentially enhance the efficacy of existing immune checkpoint therapies.
Collapse
|
85
|
Chun BM, Page DB, McArthur HL. Combination Immunotherapy Strategies in Breast Cancer. CURRENT BREAST CANCER REPORTS 2019. [DOI: 10.1007/s12609-019-00333-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
We summarize combination immunotherapy strategies for the treatment of breast cancer, with a focus on metastatic disease. First, a general overview of combination approaches is presented according to breast cancer subtype. Second, additional review of promising combination approaches is presented.
Recent Findings
Combination strategies utilizing chemotherapy or radiotherapy with immune checkpoint inhibition are being evaluated across multiple phase III trials. Dual immunotherapy strategies, such as dual immune checkpoint inhibition or combined co-stimulation/co-inhibition, have supportive preclinical evidence and are under early clinical investigation. Modulation of the immune microenvironment via cytokines and vaccination strategies, as well as locally focused treatments to enhance antigenic responses, are active areas of research.
Summary
Pre-clinical and translational research sheds new light on numerous ways the immune system may be modulated to fight against cancer. We describe current and emerging combination approaches which may improve patient outcomes in metastatic breast cancer.
Collapse
|
86
|
Gravbrot N, Gilbert-Gard K, Mehta P, Ghotmi Y, Banerjee M, Mazis C, Sundararajan S. Therapeutic Monoclonal Antibodies Targeting Immune Checkpoints for the Treatment of Solid Tumors. Antibodies (Basel) 2019; 8:E51. [PMID: 31640266 PMCID: PMC6963985 DOI: 10.3390/antib8040051] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 12/23/2022] Open
Abstract
Recently, modulation of immune checkpoints has risen to prominence as a means to treat a number of solid malignancies, given the durable response seen in many patients and improved side effect profile compared to conventional chemotherapeutic agents. Several classes of immune checkpoint modulators have been developed. Here, we review current monoclonal antibodies directed against immune checkpoints that are employed in practice today. We discuss the history, mechanism, indications, and clinical data for each class of therapies. Furthermore, we review the challenges to durable tumor responses that are seen in some patients and discuss possible interventions to circumvent these barriers.
Collapse
Affiliation(s)
- Nicholas Gravbrot
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Kacy Gilbert-Gard
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Paras Mehta
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Yarah Ghotmi
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Madhulika Banerjee
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Christopher Mazis
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
| | - Srinath Sundararajan
- Division of Hematology-Oncology, Department of Medicine, University of Arizona Cancer Center, Tucson, AZ 85724, USA.
- Texas Oncology, Dallas, TX 75251, USA.
| |
Collapse
|
87
|
Sanghera C, Sanghera R. Immunotherapy - Strategies for Expanding Its Role in the Treatment of All Major Tumor Sites. Cureus 2019; 11:e5938. [PMID: 31788395 PMCID: PMC6858270 DOI: 10.7759/cureus.5938] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Immunotherapy is widely regarded to have the ability to transform the treatment of cancer, with immune checkpoint inhibitors already in use for cancers such as advanced melanoma and non-small cell lung cancer (NSCLC). However, despite its potential, the widespread adoption of immunotherapy for the treatment of other cancers has been largely limited. This can be partly attributed to additional immunosuppressive mechanisms in the tumor microenvironment that help promote and maintain a state of T cell exhaustion. As such, the exploration of combinatory immunotherapies is an active area of research and includes the combination of immune checkpoint inhibitors with cytotoxic therapies, cancer vaccines and monoclonal antibodies against other co-inhibitory and co-stimulatory receptors. Strategies are also being employed to improve the homing, extravasation and survival of chimeric antigen receptor (CAR)-T cells in the tumor microenvironment. Furthermore, the development of immunotherapies targeted to one or multiple neoantigens unique to a specific tumor may act to enhance anti-tumor immunity, as well as reduce immune-related adverse events (irAEs). As immunotherapy evolves to become a mainstay treatment for cancer, it is imperative that optimum treatment regimens that maximize efficacy and limit toxicity are developed. Foremost, appropriate biomarkers must be identified to help tailor combinatory immunotherapies to the individual patient and hence pave the way to a new era of personalized medicine.
Collapse
Affiliation(s)
| | - Rohan Sanghera
- School of the Biological Sciences, University of Cambridge, Cambridge, GBR
| |
Collapse
|
88
|
Elaileh A, Saharia A, Potter L, Baio F, Ghafel A, Abdelrahim M, Heyne K. Promising new treatments for pancreatic cancer in the era of targeted and immune therapies. Am J Cancer Res 2019; 9:1871-1888. [PMID: 31598392 PMCID: PMC6780661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth leading cause of cancer mortality among men and women in the United States. Its incidence has been on the rise, with a projected two-fold increase by 2030. PDAC carries a poor prognosis due to a lack of effective screening tools, limited understanding of pathophysiology, and ineffective treatment modalities. Recently, there has been a revolution in the world of oncology with the advent of novel treatments to combat this disease. However, the 5-year survival of PDAC remains unchanged at a dismal 8%. The aim of this review is to bring together several studies and identify various recent modalities that have been promising in treating PDAC.
Collapse
Affiliation(s)
- Ahmed Elaileh
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Ashish Saharia
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Lucy Potter
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Flavio Baio
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Afnan Ghafel
- Department of Radiology, The University of JordanAmman, Jordan
| | - Maen Abdelrahim
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| | - Kirk Heyne
- Department of General Surgery, Houston Methodist HospitalHouston, Texas, USA
| |
Collapse
|
89
|
Abstract
Immunomodulatory antibodies that directly trigger and reawaken suppressed T-cell effector function are termed 'checkpoint inhibitors'. CTLA-4 and PD-1/PD-L1 molecules are the most studied inhibitory immune check points against cancer and because of this therapeutic property have entered the clinic for treating a variety of tumor types. The results so far demonstrate a positive impact on cancer remission. Preclinical studies have demonstrated that targeting a number of other T-cell surface molecules including both positive and negative immune regulators, also possesses strong antitumor activity. Some of these molecules have already entered clinical trials. In this report, we briefly highlight the status of these immune checkpoint inhibitors and discuss their side effects and future directions for their use.
Collapse
Affiliation(s)
- Dass S Vinay
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Byoung S Kwon
- Section of Clinical Immunology, Allergy & Rheumatology, School of Medicine, Tulane University, New Orleans, LA 70112, USA.,Eutilex Institute for Biomedical Research, Suite #1401 Daeryung Technotown 17, Gasan digital 1-ro 25, Geumcheon-gu, Seoul Korea
| |
Collapse
|
90
|
DeLeon TT, Zhou Y, Nagalo BM, Yokoda RT, Ahn DH, Ramanathan RK, Salomao MA, Aqel BA, Mahipal A, Bekaii-Saab TS, Borad MJ. Novel immunotherapy strategies for hepatobiliary cancers. Immunotherapy 2019; 10:1077-1091. [PMID: 30185133 DOI: 10.2217/imt-2018-0024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Despite recent advancements in therapeutic options for advanced hepatobiliary cancers, there remains an unmet need for innovative systemic treatments. Immunotherapy has shown an ability to provide prolonged clinical benefit, but this benefit remains limited to a small subset of patients. Numerous ongoing endeavors are investigating novel immunotherapy concepts. Immunotherapies that have demonstrated clinical efficacy in hepatobiliary cancers include PD-1 inhibitor therapy and CTLA-4 inhibitor therapy. Novel immunotherapy concepts include targeting emerging checkpoint proteins, bispecific T-cell engagers, combinatorial trials with checkpoint inhibitors, oncolytic virotherapy and chimeric antigen receptor T cells. The goal for these new treatment strategies is to achieve a meaningful expansion of patients deriving prolonged clinical benefit from immunotherapy.
Collapse
Affiliation(s)
- Thomas T DeLeon
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Yumei Zhou
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Bolni M Nagalo
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Raquel T Yokoda
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Daniel H Ahn
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Ramesh K Ramanathan
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Marcela A Salomao
- Department of Pathology, Division of Anatomic Pathology & Laboratory Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Bashar A Aqel
- Department of Medicine, Division of Gastroenterology & Hepatology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Amit Mahipal
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Tanios S Bekaii-Saab
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Mitesh J Borad
- Department of Medicine, Division of Hematology & Oncology, Mayo Clinic, Scottsdale, AZ 85259, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Mayo Clinic Cancer Center, Phoenix, AZ 85054, USA
| |
Collapse
|
91
|
Nanotechnology is an important strategy for combinational innovative chemo-immunotherapies against colorectal cancer. J Control Release 2019; 307:108-138. [DOI: 10.1016/j.jconrel.2019.06.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 12/15/2022]
|
92
|
Kochin V, Nishikawa H. <Editors' Choice> Meddling with meddlers: curbing regulatory T cells and augmenting antitumor immunity. NAGOYA JOURNAL OF MEDICAL SCIENCE 2019; 81:1-18. [PMID: 30962651 PMCID: PMC6433633 DOI: 10.18999/nagjms.81.1.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
CD4+ regulatory T cells (Tregs) expressing the transcription factor forkhead
box P3 (FoxP3) play an important role in self-tolerance and immune homeostasis. Tregs have
evolved to protect the host from aberrant immune responses against self-components and
collateral damages occurring in the process of defense against invading pathogens by
softening immune responses. However, they turned to be a scourge in malignant tumors by
not only allowing and promoting tumor growth but also suppressing effective antitumor
actions, both inherent (host’s immune surveillance) and extrinsic (anticancer therapy). An
increase in the number of Tregs infiltrating into tumor sites and a concomitant decrease
in the number of CD8+ cytotoxic T lymphocytes are associated with a poor
prognosis for various types of cancers, marking Tregs as notorious meddlers with an
effective antitumor response. Various cancer immunotherapy approaches are often dampened
by meddling Tregs, making them one of the major targets in the treatment of cancer. The
recent success of immune checkpoint inhibitors (ICIs) that target immune checkpoint
molecules expressed by Tregs or effector T cells implies, that “meddling with meddlers”
represents an effective strategy in cancer immunotherapy. However, clinical responses to
ICIs are effective and durable only in some patients with cancer, whereas more than half
of them do not show significant clinical improvement. This implies that a therapeutic
approach based on the use of a single ICI, or targeting Tregs alone, is insufficient,
highlighting the need for combinatorial approaches. With regard to antitumor immune
stimulation, several approaches, such as vaccination with peptides (or the corresponding
DNA) to stimulate antigen-presenting CD8+ T cells with tumor-specific
neoantigens, cancer/testis antigens, or cancer stem cell antigens, that eventually boost
effective cytotoxic antitumor responses are being tested. This review describes the
immunosuppressive physiology of Tregs and their meddling with the host’s antitumor
immunity; current and prospective approaches to curb Tregs; and approaches to augment
antitumor immunity.
Collapse
Affiliation(s)
- Vitaly Kochin
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroyoshi Nishikawa
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo / Chiba, Japan
| |
Collapse
|
93
|
Harrington K, Freeman DJ, Kelly B, Harper J, Soria JC. Optimizing oncolytic virotherapy in cancer treatment. Nat Rev Drug Discov 2019; 18:689-706. [PMID: 31292532 DOI: 10.1038/s41573-019-0029-0] [Citation(s) in RCA: 309] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2019] [Indexed: 02/07/2023]
Abstract
In the wake of the success of modern immunotherapy, oncolytic viruses (OVs) are currently seen as a potential therapeutic option for patients with cancer who do not respond or fail to achieve durable responses following treatment with immune checkpoint inhibitors. OVs offer a multifaceted therapeutic platform because they preferentially replicate in tumour cells, can be engineered to express transgenes that augment their cytotoxic and immunostimulatory activities, and modulate the tumour microenvironment to optimize immune-mediated tumour eradication, both at locoregional and systemic sites of disease. Lysis of tumour cells releases tumour-specific antigens that trigger both the innate and adaptive immune systems. OVs also represent attractive combination partners with other systemically delivered agents by virtue of their highly favourable safety profiles. Rational combinations of OVs with different immune modifiers and/or antitumour agents, based on mechanisms of tumour resistance to immune-mediated attack, may benefit the large, currently underserved, population of patients who respond poorly to immune checkpoint inhibition.
Collapse
Affiliation(s)
- Kevin Harrington
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK.
| | | | - Beth Kelly
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | - Jean-Charles Soria
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA.,Department of Medicine and Medical Oncology, Université Paris-Sud, Orsay, France
| |
Collapse
|
94
|
Ciardiello D, Vitiello PP, Cardone C, Martini G, Troiani T, Martinelli E, Ciardiello F. Immunotherapy of colorectal cancer: Challenges for therapeutic efficacy. Cancer Treat Rev 2019; 76:22-32. [DOI: 10.1016/j.ctrv.2019.04.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/12/2019] [Accepted: 04/15/2019] [Indexed: 02/06/2023]
|
95
|
Jacobi FJ, Wild K, Smits M, Zoldan K, Csernalabics B, Flecken T, Lang J, Ehrenmann P, Emmerich F, Hofmann M, Thimme R, Neumann-Haefelin C, Boettler T. OX40 stimulation and PD-L1 blockade synergistically augment HBV-specific CD4 T cells in patients with HBeAg-negative infection. J Hepatol 2019; 70:1103-1113. [PMID: 30826436 DOI: 10.1016/j.jhep.2019.02.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Current antiviral therapies lack the potential to eliminate persistent hepatitis B virus (HBV) infection. HBV-specific T cells are crucial for HBV control and have recently been shown to be protective in patients following discontinuation of antiviral therapy. Thus, T cell-based approaches may greatly improve the therapeutic landscape of HBV infection. We aimed to augment HBV-specific CD4 T cells from chronically infected patients by targeting different immunological pathways. METHODS Expression of various co-stimulatory and inhibitory receptors on HBV- and influenza-specific CD4 T cells was analyzed directly ex vivo by MHC class II-tetramers. Patients infected with HBV genotype D were screened for CD4 T cell responses by IFN-γ ELISpot and intracellular cytokine staining following stimulation with overlapping peptides (OLPs) spanning the HBV-polyprotein. Stimulation with recombinant IL-7, an agonistic OX40-antibody or blockade of PD-L1 was performed in antigen-specific in vitro cultures. Cytokine secretion and expression of transcription factors were analyzed by flow cytometry. Responses targeting influenza, Epstein-Barr virus and tetanus toxoid served as controls. RESULTS Tetramer-staining revealed that the IL-7 receptor-alpha (CD127), OX40 and PD-1 constitute possible therapeutic targets as they were all strongly expressed on HBV-specific CD4 T cells ex vivo. The HBV-specific CD4 T cell responses identified by OLP screening targeted predominantly the HBV-polymerase and core proteins. Combined OX40 stimulation and PD-L1 blockade significantly augmented IFN-γ and IL-21 producing HBV-specific CD4 T cells in vitro, suggesting active T helper type 1 cell and follicular T helper cell programs. Indeed, transcription factors T-bet and Bcl6 were strongly expressed in cytokine-producing cells. CONCLUSIONS Combined OX40 stimulation and PD-L1 blockade augmented secretion of the helper T cell signature cytokines IFN-γ and IL-21, suggesting that immunotherapeutic approaches can improve HBV-specific CD4 T cell responses. LAY SUMMARY CD4 T cells are important in controlling viral infections but are impaired in the context of chronic hepatitis B virus (HBV) infection. Therapeutic approaches to cure chronic HBV infection are highly likely to require an immune-stimulatory component. This study demonstrates that HBV-specific CD4 T cells can be functionally augmented by combined stimulation of the co-stimulatory molecule OX40 and blockade of the inhibitory PD-1 pathway.
Collapse
Affiliation(s)
- Felix Johannes Jacobi
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Katharina Wild
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Chemistry and Pharmacy, University of Freiburg, Germany
| | - Maike Smits
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany; Faculty of Biology, University of Freiburg, Germany
| | - Katharina Zoldan
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Benedikt Csernalabics
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias Flecken
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Julia Lang
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Philipp Ehrenmann
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Florian Emmerich
- Institute for Transfusion Medicine and Gene Therapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Christoph Neumann-Haefelin
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany
| | - Tobias Boettler
- Department of Medicine II, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
96
|
Mason R, Au L, Ingles Garces A, Larkin J. Current and emerging systemic therapies for cutaneous metastatic melanoma. Expert Opin Pharmacother 2019; 20:1135-1152. [PMID: 31025594 DOI: 10.1080/14656566.2019.1601700] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/27/2019] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Melanoma therapies have evolved rapidly, and initial successes have translated into survival gains for patients with advanced melanoma. Both targeted and immune-therapy now have evidence in earlier stage disease. There are many new agents and combinations of treatments in development as potential future treatment options. This highlights the need for a reflection on current treatment practice trends that are guiding the development of potential new therapies. AREAS COVERED In this review, the authors discuss the evidence for currently approved therapies for cutaneous melanoma, including adjuvant therapy, potential new biomarkers, and emerging treatments with early phase clinical trial data. The authors have searched both the PubMed and clinicaltrials.gov databases for published clinical trials and discuss selected landmark trials of current therapies and of investigational treatment strategies with early evidence for the treatment of melanoma. EXPERT OPINION Significant efficacy has been demonstrated with both immune checkpoint inhibitors and targeted therapies in treating advanced melanoma. A multitude of novel therapies are in development and there is need for instructive biomarker assessment to identify patients likely to respond or be refractory to current therapies, to identify mechanisms of resistance and to direct further treatment options to patients based on individual disease biology.
Collapse
Affiliation(s)
- Robert Mason
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- b Department of Medical Oncology , Gold Coast University Hospital , Southport , Queensland , Australia
| | - Lewis Au
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
| | - Alvaro Ingles Garces
- a Clinical Research Fellow, Skin and Renal Units , The Royal Marsden Hospital , London , UK
| | - James Larkin
- c Division of Clinical Research , The Institute of Cancer Research , London , UK
- d Consultant Oncologist , The Royal Marsden Hospital , London , UK
| |
Collapse
|
97
|
Khan IA, Hwang S, Moretto M. Toxoplasma gondii: CD8 T Cells Cry for CD4 Help. Front Cell Infect Microbiol 2019; 9:136. [PMID: 31119107 PMCID: PMC6504686 DOI: 10.3389/fcimb.2019.00136] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/15/2019] [Indexed: 11/13/2022] Open
Abstract
Toxoplasma gondii, an apicomplexan parasite, is a pathogenic protozoan that can infect the central nervous system. In pregnant women, infection can result in congenital problems of the fetus, while in immunocompromised individual it can lead to severe neurological consequences. Although CD8 T cells play an important effector role in controlling the chronic infection, their maintenance is dependent on the critical help provided by CD4 T cells. In a recent study, we demonstrated that reactivation of the infection in chronically infected host is a consequence of CD8 T dysfunction caused by CD4 T cell exhaustion. Furthermore, treatment of chronically infected host with antigen-specific non-exhausted CD4 T cells can restore CD8 T cell functionality and prevent reactivation of the latent infection. The exhaustion status of CD4 T cells is mediated by the increased expression of the transcription factor BLIMP-1, and deletion of this molecule led to the restoration of CD4 T cell function, reversal of CD8 exhaustion and prevention of reactivation of the latent infection. In a recent study from our laboratory, we also observed an increased expression of miR146a levels by CD4 T cells from the chronically infected animals. Recent reports have demonstrated that microRNAs (especially miR146a) has a strong impact on the immune system of T. gondii infected host. Whether these molecules have any role in the BLIMP-1 up-regulation and dysfunctionality of these cells needs to be investigated.
Collapse
Affiliation(s)
- Imtiaz A. Khan
- Department Microbiology, Immunology and Tropical Medicine, The George Washington University, Washington, DC, United States
| | | | | |
Collapse
|
98
|
Philipp Bewersdorf J, Stahl M, Zeidan AM. Immune checkpoint-based therapy in myeloid malignancies: a promise yet to be fulfilled. Expert Rev Anticancer Ther 2019; 19:393-404. [PMID: 30887841 PMCID: PMC6527485 DOI: 10.1080/14737140.2019.1589374] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 02/27/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Immune system evasion is essential for tumor cell survival and is mediated by the immunosuppressive tumor microenvironment and the activation of inhibitory immune checkpoints. While immune checkpoint-based therapy yielded impressive results in several advanced solid malignancies such as melanoma and non-small cell lung cancer, its role in acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) is still evolving. Areas covered: Here we review the immunology in the tumor microenvironment in the bone marrow and discuss the current preclinical and clinical data for immune checkpoint-based therapy in myeloid neoplasms. Expert commentary: Clinical trials of immune checkpoint inhibitors (ICI) in AML and MDS are still in early stages and reported results so far have been modest especially for monotherapy use in the refractory settings. However, there are preliminary data for synergistic effects for combination of multiple ICI with hypomethylating agents and conventional chemotherapy. ICI might also be effective in eradicating minimal residual disease and to prevent relapse following induction chemotherapy or hematopoietic stem cell transplant. Additional trials to provide insight into the efficacy and safety profile of immune checkpoint-based therapy, its optimal timing and potential combination with other types of therapy as well as identification of predictive biomarkers are needed.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| | - Maximilian Stahl
- Department of Medicine, Section of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
99
|
Beha N, Harder M, Ring S, Kontermann RE, Müller D. IL15-Based Trifunctional Antibody-Fusion Proteins with Costimulatory TNF-Superfamily Ligands in the Single-Chain Format for Cancer Immunotherapy. Mol Cancer Ther 2019; 18:1278-1288. [PMID: 31040163 DOI: 10.1158/1535-7163.mct-18-1204] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/08/2019] [Accepted: 04/25/2019] [Indexed: 11/16/2022]
Abstract
IL15 and costimulatory receptors of the tumor necrosis superfamily (TNFRSF) have shown great potential to support and drive an antitumor immune response. However, their efficacy as monotherapy is limited. Here, we present the development of a novel format for a trifunctional antibody-fusion protein that combines and focuses the activity of IL15/TNFSF-ligand in a targeting-mediated manner to the tumor site. The previously reported format consisted of a tumor-directed antibody (scFv), IL15 linked to an IL15Rα-fragment (RD), and the extracellular domain of 4-1BBL, where noncovalent trimerization of 4-1BBL into its functional unit led to a homotrimeric molecule with 3 antibody and 3 IL15-RD units. To reduce the size and complexity of the molecule, we have now designed a second format, where 4-1BBL is introduced as single-chain (sc), that is 3 consecutively linked 4-1BBL ectodomains. Thus, a monomeric trifunctional fusion protein presenting only 1 functional unit of each component was generated. Interestingly, the in vitro activity on T-cell stimulation was conserved or even enhanced for the soluble and target-bound molecule, respectively. Also, in a lung tumor mouse model, comparable antitumor effects were observed. Furthermore, corroborating the concept, OX40L and GITRL were also successfully incorporated into the novel single-chain format and the advantage of target-bound trifunctional versus corresponding combined bifunctional fusion proteins demonstrated by measuring T-cell proliferation and cytotoxic potential in vitro and antitumor effects of RD_IL15_scFv_scGITRL in a lung tumor mouse model in vivo Thus, the trifunctional antibody-fusion protein single-chain format constitutes a promising innovative platform for further therapeutic developments.
Collapse
Affiliation(s)
- Nadine Beha
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Markus Harder
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sarah Ring
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Dafne Müller
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.
| |
Collapse
|
100
|
Medina-Cucurella AV, Mizrahi RA, Asensio MA, Edgar RC, Leong J, Leong R, Lim YW, Nelson A, Niedecken AR, Simons JF, Spindler MJ, Stadtmiller K, Wayham N, Adler AS, Johnson DS. Preferential Identification of Agonistic OX40 Antibodies by Using Cell Lysate to Pan Natively Paired, Humanized Mouse-Derived Yeast Surface Display Libraries. Antibodies (Basel) 2019; 8:antib8010017. [PMID: 31544823 PMCID: PMC6640694 DOI: 10.3390/antib8010017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 12/18/2022] Open
Abstract
To discover therapeutically relevant antibody candidates, many groups use mouse immunization followed by hybridoma generation or B cell screening. One modern approach is to screen B cells by generating natively paired single chain variable fragment (scFv) display libraries in yeast. Such methods typically rely on soluble antigens for scFv library screening. However, many therapeutically relevant cell-surface targets are difficult to express in a soluble protein format, complicating discovery. In this study, we developed methods to screen humanized mouse-derived yeast scFv libraries using recombinant OX40 protein in cell lysate. We used deep sequencing to compare screening with cell lysate to screening with soluble OX40 protein, in the context of mouse immunizations using either soluble OX40 or OX40-expressing cells and OX40-encoding DNA vector. We found that all tested methods produce a unique diversity of scFv binders. However, when we reformatted forty-one of these scFv as full-length monoclonal antibodies (mAbs), we observed that mAbs identified using soluble antigen immunization with cell lysate sorting always bound cell surface OX40, whereas other methods had significant false positive rates. Antibodies identified using soluble antigen immunization and cell lysate sorting were also significantly more likely to activate OX40 in a cellular assay. Our data suggest that sorting with OX40 protein in cell lysate is more likely than other methods to retain the epitopes required for antibody-mediated OX40 agonism.
Collapse
Affiliation(s)
- Angélica V Medina-Cucurella
- Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824, USA.
| | - Rena A Mizrahi
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Michael A Asensio
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Robert C Edgar
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Jackson Leong
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Renee Leong
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Yoong Wearn Lim
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Ayla Nelson
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Ariel R Niedecken
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Jan Fredrik Simons
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Matthew J Spindler
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Kacy Stadtmiller
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Nicholas Wayham
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - Adam S Adler
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| | - David S Johnson
- GigaGen Inc., One Tower Place, Suite 750, South San Francisco, CA 94080, USA.
| |
Collapse
|