1
|
Ye H, Liao W, Pan J, Shi Y, Wang Q. Immune checkpoint blockade for cancer therapy: current progress and perspectives. J Zhejiang Univ Sci B 2025; 26:203-226. [PMID: 40082201 PMCID: PMC11906392 DOI: 10.1631/jzus.b2300492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 12/05/2023] [Indexed: 03/16/2025]
Abstract
Dysfunction of anti-tumor immune responses is crucial for cancer progression. Immune checkpoint blockade (ICB), which can potentiate T cell responses, is an effective strategy for the normalization of host anti-tumor immunity. In recent years, immune checkpoints, expressed on both tumor cells and immune cells, have been identified; some of them have exhibited potential druggability and have been approved by the US Food and Drug Administration (FDA) for clinical treatment. However, limited responses and immune-related adverse events (irAEs) cannot be ignored. This review outlines the development and applications of ICBs, potential strategies for overcoming resistance, and future directions for ICB-based cancer immunotherapy.
Collapse
Affiliation(s)
- Hongying Ye
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Weijie Liao
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Jiongli Pan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China
| | - Yin Shi
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China.
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou 310058, China.
| |
Collapse
|
2
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 PMCID: PMC11874808 DOI: 10.1186/s12951-025-03236-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
3
|
Ignatiadis M, Bailey A, McArthur H, El-abed S, de Azambuja E, Metzger O, Chui SY, Dieterich M, Perretti T, Shearer-Kang E, Molinero L, Steger GG, Jassem J, Lee SC, Higgins M, Zarba J, Schmidt M, Gomez H, Guerrero Zotano A, Moscetti L, Chiu J, Munzone E, Ben-Baruch NE, Bajetta E, Ohno S, Im SA, Werutsky G, Gal-Yam EN, Gonzalez Farre X, Tseng LM, Jacot W, Gluz O, Shao Z, Shparyk Y, Zimina A, Winer E, Cameron DA, Viale G, Saji S, Gelber R, Piccart M. Adjuvant Atezolizumab for Early Triple-Negative Breast Cancer: The ALEXANDRA/IMpassion030 Randomized Clinical Trial. JAMA 2025:2829800. [PMID: 39883436 PMCID: PMC11783246 DOI: 10.1001/jama.2024.26886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/27/2024] [Indexed: 01/31/2025]
Abstract
Importance Triple-negative breast cancer is an aggressive subtype with a high incidence in young patients, a high incidence in non-Hispanic Black women, and a high risk of progression to metastatic cancer, a devastating sequela with a 12- to 18-month life expectancy. Until recently, one strategy for treating early-stage triple-negative breast cancer was chemotherapy after surgery. However, it was not known whether the addition of immune therapy to postsurgery chemotherapy would be beneficial. Objective To evaluate the addition of immune therapy in the form of atezolizumab to postoperative chemotherapy in patients with the high-risk triple-negative breast cancer subtype. Design, Setting, and Participants In this open-label international randomized phase 3 trial conducted in more than 330 centers in 31 countries, patients undergoing surgery as initial treatment for stage II or III triple-negative breast cancer were enrolled between August 2, 2018, and November 11, 2022. The last patient follow-up was on August 18, 2023. Interventions Patients were randomized (1:1) to receive standard chemotherapy for 20 weeks with (n = 1101) or without (n = 1098) the immune therapy drug atezolizumab for up to 1 year. Main Outcomes and Measures The primary end point was invasive disease-free survival (time between randomization and invasive breast cancer in the same or opposite breast, recurrence elsewhere in the body, or death from any cause). Results The median age of enrolled patients was 53 years and most self-reported as being of Asian or White race and neither Latino nor Hispanic ethnicity. The study independent data monitoring committee halted enrollment at 2199 of 2300 planned patients. All patients stopped atezolizumab following a planned early interim and futility analysis. The trial continued to a premature final analysis. With invasive disease-free survival events in 141 patients (12.8%) treated with atezolizumab-chemotherapy and 125 (11.4%) with chemotherapy alone (median follow-up, 32 months), the final stratified invasive disease-free survival hazard ratio was 1.11 (95% CI, 0.87-1.42; P = .38). Compared with chemotherapy alone, the regimen of atezolizumab plus chemotherapy was associated with more treatment-related grade 3 or 4 adverse events (54% vs 44%) but similar incidences of fatal adverse events (0.8% vs 0.6%) and adverse events leading to chemotherapy discontinuation. Chemotherapy exposure was similar in the 2 treatment groups. Conclusions and Relevance The addition of the immune therapy drug atezolizumab to chemotherapy after surgery did not provide benefit among patients with triple-negative breast cancer who are at high risk of recurrent disease. Trial Registration ClinicalTrials.gov Identifier: NCT03498716.
Collapse
Affiliation(s)
- Michail Ignatiadis
- Institut Jules Bordet, l’Université Libre de Bruxelles and Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | | | - Heather McArthur
- Simmons Cancer Center at UT Southwestern Medical Center, Dallas, Texas
| | | | - Evandro de Azambuja
- Institut Jules Bordet, l’Université Libre de Bruxelles and Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Otto Metzger
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | | | | | | | | | | | | | - Soo Chin Lee
- National University Hospital Singapore, Singapore
| | | | - Jose Zarba
- National University of Tucaman, Tucaman, Argentina
| | - Marcus Schmidt
- Comprehensive Cancer Center University Medical Center Mainz, Mainz, Germany
| | - Henry Gomez
- National Institute of Neoplastic Diseases, Lima, Peru
| | - Angel Guerrero Zotano
- Oncology Institute, Valencia, Spain
- Grupo Español de Investigación en Cáncer de Mama, Madrid, Spain
| | | | - Joanne Chiu
- Queen Mary Hospital & Gleneagles Hospital Hong Kong, Hong Kong
| | | | | | | | - Shinji Ohno
- Cancer Institute of the Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seock-Ah Im
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Oleg Gluz
- Breast Center Niederrhein, Mönchengladbach, Germany
| | - Zhimin Shao
- Fudan University Cancer Institute, Shanghai, China
| | | | | | - Eric Winer
- Yale Cancer Center, New Haven, Connecticut
| | - David A. Cameron
- Breast International Group, Brussels, Belgium
- University of Edinburgh, Edinburgh, United Kingdom
| | | | | | - Richard Gelber
- Dana-Farber Cancer Institute, Boston, Massachusetts
- Frontier Science, Boston, Massachusetts
| | - Martine Piccart
- Institut Jules Bordet, l’Université Libre de Bruxelles and Hôpital Universitaire de Bruxelles, Brussels, Belgium
| |
Collapse
|
4
|
Gallego RA, Cho-Schultz S, Del Bel M, Dechert-Schmitt AM, Donaldson JS, He M, Jalaie M, Kania R, Matthews J, McTigue M, Tuttle JB, Risley H, Zhou D, Zhou R, Ahmad OK, Bernier L, Berritt S, Braganza J, Chen Z, Cianfrogna JA, Collins M, Costa Jones C, Cronin CN, Davis C, Dress K, Edwards M, Farrell W, France SP, Grable N, Johnson E, Johnson TW, Jones R, Knauber T, Lafontaine J, Loach RP, Maestre M, Miller N, Moen M, Monfette S, Morse P, Nager AR, Niosi M, Richardson P, Rohner AK, Sach NW, Timofeevski S, Tucker JW, Vetelino B, Zhang L, Nair SK. Discovery of PF-07265028, A Selective Small Molecule Inhibitor of Hematopoietic Progenitor Kinase 1 (HPK1) for the Treatment of Cancer. J Med Chem 2024; 67:22002-22038. [PMID: 39651809 DOI: 10.1021/acs.jmedchem.4c01930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hematopoietic progenitor kinase 1 (HPK1/MAP4K1) represents a high interest target for the treatment of cancer through an immune-mediated mechanism. Herein we present highlights of the drug discovery campaign within the lactam/azalactam series of inhibitors that yielded a small molecule (21, PF-07265028), which was advanced to a phase 1 clinical trial (NCT05233436). Key components of the discovery effort included optimization of potency through mitigation of ligand strain as guided by the use of cocrystal structures, mitigation of ADME liabilities (plasma instability and fraction metabolism by CYP2D6), and optimization of kinase selectivity, particularly over immune-modulating kinases with high homology to HPK1. Structure-based drug design via leveraging cocrystal structures and lipophilic efficiency analysis proved to be valuable tools that ultimately enabled the delivery of a clinical-quality small molecule inhibitor of HPK1.
Collapse
Affiliation(s)
- Rebecca A Gallego
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sujin Cho-Schultz
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Matthew Del Bel
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | | | - Joyann S Donaldson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mingying He
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mehran Jalaie
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Rob Kania
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jean Matthews
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michele McTigue
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jamison B Tuttle
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Hud Risley
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Dahui Zhou
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Ru Zhou
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Omar K Ahmad
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Louise Bernier
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Simon Berritt
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - John Braganza
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Zecheng Chen
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Julie A Cianfrogna
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michael Collins
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Cinthia Costa Jones
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ciaran N Cronin
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Carl Davis
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Klaus Dress
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Martin Edwards
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - William Farrell
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Scott P France
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Nicole Grable
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Eric Johnson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ted W Johnson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Rhys Jones
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Thomas Knauber
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Jennifer Lafontaine
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Richard P Loach
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Michael Maestre
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Nichol Miller
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mark Moen
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Sebastien Monfette
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Peter Morse
- Pharmacokinetics, Dynamics and Metabolism Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Andrew Ross Nager
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Mark Niosi
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Paul Richardson
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Allison K Rohner
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Neal W Sach
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sergei Timofeevski
- Oncology Research Unit Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Joseph W Tucker
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Beth Vetelino
- Worldwide Research and Development, Pfizer, Inc., Groton, Connecticut 06340, United States
| | - Lei Zhang
- Worldwide Research and Development, Pfizer, Inc., Cambridge, Massachusetts 02139, United States
| | - Sajiv K Nair
- Oncology Medicinal Chemistry Worldwide Research and Development, Pfizer, Inc., 10770 Science Center Drive, La Jolla, California 92121, United States
| |
Collapse
|
5
|
Li J, Hu X, Zhang S. Efficacy and side effects of pembrolizumab plus chemotherapy vs. chemotherapy alone in patients with advanced gastric or gastroesophageal junction adenocarcinoma: A meta‑analysis. Oncol Lett 2024; 28:371. [PMID: 38910906 PMCID: PMC11190732 DOI: 10.3892/ol.2024.14504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
Recently, the treatment plan of pembrolizumab plus chemotherapy was regarded as a promising treatment for patients with advanced gastric cancer or gastroesophageal junction adenocarcinoma (GC/GEJC). However, the efficacy and side effects of pembrolizumab plus chemotherapy still lack evidence-based medical evidence to support. Therefore, a meta-analysis was conducted to evaluate the hot issue. By searching PubMed, EMBASE, Cochrane Library, Web of Science, any randomized clinical studies of pembrolizumab plus chemotherapy versus chemotherapy in patients with advanced GC/GEJC met the inclusion criteria were included. The quality of the literature was evaluated and the data was extracted. A correlative software was also used to analyze the data and to draw a conclusion. After screening 14,015 studies, four studies were eligible for the meta-analysis. Compared with chemotherapy alone group, the overall survival (OS) rate was significantly longer. In programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥1 subgroup and PD-L1 CPS ≥10 subgroup analyses, the results showed that the response rate (RR) and complete response rate (CR) were both higher in pembrolizumab plus chemotherapy group compared with chemotherapy alone group. There were not significant differences in the CR, the treatment-related adverse events, succumbed to drug-related events and succumbed to immune-mediated events between the two groups. However, the effect events such as the treatment-related adverse events led to discontinuation, the 3-5 treatment-related adverse events and the immune-mediated adverse events and infusion reactions were more common in pembrolizumab plus chemotherapy group. In conclusion, the current meta-analysis revealed that, in treating advanced GC/GEJC, pembrolizumab plus chemotherapy had improved therapeutic efficacies than chemotherapy alone, as evidenced by the significantly longer OS. Furthermore, the patients in PD-L1 CPS ≥1 subgroup and PD-L1 CPS ≥10 subgroup appeared to benefit from pembrolizumab plus chemotherapy treatment because of higher RR and CR. However, side effects such as the treatment-related adverse events leading to discontinuation, the 3-5 treatment-related adverse events, and immune-mediated adverse events and infusion reactions deserved more attention.
Collapse
Affiliation(s)
- Jinquan Li
- Department of Gastrointestinal Surgery, The First People's Hospital of Jingdezhen, Jingdezhen, Jiangxi 33300, P.R. China
| | - Xiaosheng Hu
- Department of Gastrointestinal Surgery, The First People's Hospital of Jingdezhen, Jingdezhen, Jiangxi 33300, P.R. China
| | - Shanzhong Zhang
- Department of Gastrointestinal Surgery, The First People's Hospital of Jingdezhen, Jingdezhen, Jiangxi 33300, P.R. China
| |
Collapse
|
6
|
Sui Y, Berzofsky JA. Trained immunity inducers in cancer immunotherapy. Front Immunol 2024; 15:1427443. [PMID: 39081326 PMCID: PMC11286386 DOI: 10.3389/fimmu.2024.1427443] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
While most of the cancer immunotherapy strategies engage adaptive immunity, especially tumor-associated T cells, the small fraction of responding patients and types of cancers amenable, and the possibility of severe adverse effects limit its usage. More effective and general interventions are urgently needed. Recently, a de facto innate immune memory, termed 'trained immunity', has become a new research focal point, and promises to be a powerful tool for achieving long-term therapeutic benefits against cancers. Trained immunity-inducing agents such as BCG and fungal glucan have been shown to be able to avert the suppressive tumor microenvironment (TME), enhance T cell responses, and eventually lead to tumor regression. Here, we review the current understating of trained immunity induction and highlight the critical roles of emergency granulopoiesis, interferon γ and tissue-specific induction. Preclinical and clinical studies that have exploited trained immunity inducers for cancer immunotherapy are summarized, and repurposed trained immunity inducers from other fields are proposed. We also outline the challenges and opportunities for trained immunity in future cancer immunotherapies. We envisage that more effective cancer vaccines will combine the induction of trained immunity with T cell therapies.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, United States
| | | |
Collapse
|
7
|
Yoshida K, Asanuma K, Matsuyama Y, Okamoto T, Hagi T, Nakamura T, Sudo A. Release of Exosomal PD-L1 in Bone and Soft Tissue Sarcomas and Its Relationship to Radiotherapy. Cancers (Basel) 2024; 16:2489. [PMID: 39001550 PMCID: PMC11240571 DOI: 10.3390/cancers16132489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
(1) Background: Exosomal PD-L1 has garnered attention owing to its role in instigating systemic immune suppression. The objective of this study is to elucidate whether bone and soft tissue sarcoma cells possess the capacity to secrete functionally active exosomal PD-L1 and whether radiotherapy (RT) induces the exosomal PD-L1 release. (2) Methods: Human osteosarcoma cell line 143B and human fibrosarcoma cell line HT1080 were utilized. Exosomes were isolated from the culture medium and blood via ultracentrifugation. The expression of PD-L1 on both tumor cells and exosomes was evaluated. The inhibitory effect on PBMC was employed to assess the activity of exosomal PD-L1. Post radiotherapy, changes in PD-L1 expression were compared. (3) Results: Exosomal PD-L1 was detected in the culture medium of tumor cells but was absent in the culture medium of PD-L1 knockout cells. Exosomal PD-L1 exhibited an inhibitory effect on PBMC activation. In tumor-bearing mice, human-derived exosomal PD-L1 was detected in the bloodstream. Following radiotherapy, tumor cells upregulated PD-L1, and human-derived exosomal PD-L1 were detected in the bloodstream. (4) Conclusions: Exosomal PD-L1 emanates from bone and soft tissue sarcoma cells and is disseminated into the circulatory system. The levels of PD-L1 in tumor cells and the release of exosomal PD-L1 were augmented after irradiation with RT.
Collapse
Affiliation(s)
- Keisuke Yoshida
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Kunihiro Asanuma
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Yumi Matsuyama
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Takayuki Okamoto
- Department of Pharmacology, Faculty of Medicine, Shimane University, Izumo 693-0021, Shimane, Japan
| | - Tomohito Hagi
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Tomoki Nakamura
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu 514-8507, Mie, Japan
| |
Collapse
|
8
|
Li B, Zu M, Jiang A, Cao Y, Wu J, Shahbazi MA, Shi X, Reis RL, Kundu SC, Xiao B. Magnetic natural lipid nanoparticles for oral treatment of colorectal cancer through potentiated antitumor immunity and microbiota metabolite regulation. Biomaterials 2024; 307:122530. [PMID: 38493672 DOI: 10.1016/j.biomaterials.2024.122530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/10/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
The therapeutic efficacy of oral nanotherapeutics against colorectal cancer (CRC) is restricted by inadequate drug accumulation, immunosuppressive microenvironment, and intestinal microbiota imbalance. To overcome these challenges, we elaborately constructed 6-gingerol (Gin)-loaded magnetic mesoporous silicon nanoparticles and functionalized their surface with mulberry leaf-extracted lipids (MLLs) and Pluronic F127 (P127). In vitro experiments revealed that P127 functionalization and alternating magnetic fields (AMFs) promoted internalization of the obtained P127-MLL@Gins by colorectal tumor cells and induced their apoptosis/ferroptosis through Gin/ferrous ion-induced oxidative stress and magneto-thermal effect. After oral administration, P127-MLL@Gins safely passed to the colorectal lumen, infiltrated the mucus barrier, and penetrated into the deep tumors under the influence of AMFs. Subsequently, the P127-MLL@Gin (+ AMF) treatment activated antitumor immunity and suppressed tumor growth. We also found that this therapeutic modality significantly increased the abundance of beneficial bacteria (e.g., Bacillus and unclassified-c-Bacilli), reduced the proportions of harmful bacteria (e.g., Bacteroides and Alloprevotella), and increased lipid oxidation metabolites. Strikingly, checkpoint blockers synergistically improved the therapeutic outcomes of P127-MLL@Gins (+ AMF) against orthotopic and distant colorectal tumors and significantly prolonged mouse life spans. Overall, this oral therapeutic platform is a promising modality for synergistic treatment of CRC.
Collapse
Affiliation(s)
- Baoyi Li
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Menghang Zu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Aodi Jiang
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Yingui Cao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Jiaxue Wu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, Netherlands
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães, 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, 4800-058, Portugal
| | - Bo Xiao
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
9
|
Huang J, Liu X, Lin M, Xiao Z, Shuai X. Light-inducible nanodrug-mediated photodynamic and anti-apoptotic synergy for enhanced immunotherapy in triple-negative breast cancer. Biomater Sci 2024; 12:2639-2647. [PMID: 38563394 DOI: 10.1039/d4bm00083h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Triple negative breast cancer (TNBC) exhibits limited responsiveness to immunotherapy owing to its immunosuppressive tumor microenvironment (TME). Here, a reactive oxygen species (ROS)-labile nanodrug encapsulating the photosensitizer Ce6 and Bcl-2 inhibitor ABT-737 was developed to provoke a robust immune response via the synergistic effect of photodynamic therapy (PDT) and the reversal of apoptosis resistance. Upon exposure to first-wave near-infrared laser irradiation, the generated ROS triggers PEG cleavage, facilitating the accumulation of the nanodrug at tumor region and endocytosis by tumor cells. Further irradiation leads to the substantial generation of cytotoxic ROS, initiating an immunogenic cell death (ICD) cascade, which prompts the maturation of dendritic cells (DCs) as well as the infiltration of T cells into the tumor site. Meanwhile, Bcl-2 inhibition counteracts apoptosis resistance, thereby amplifying PDT-induced ICD and bolstering antitumor immunity. As a result, the ROS-sensitive nanodrug demonstrates a potent inhibitory effect on tumor growth.
Collapse
Affiliation(s)
- Jing Huang
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, P. R. China.
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, P. R. China
- School of Engineering, Westlake University, Hangzhou 310030, P. R. China
| | - Xingliang Liu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, P. R. China.
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, P. R. China
- School of Engineering, Westlake University, Hangzhou 310030, P. R. China
| | - Minzhao Lin
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, P. R. China
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, P. R. China.
| | - Xintao Shuai
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, P. R. China.
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, P. R. China
| |
Collapse
|
10
|
Liu M, Jin D, Yu W, Yu J, Cao K, Cheng J, Zheng X, Wang A, Liu Y. Enhancing Tumor Immunotherapy by Multivalent Anti-PD-L1 Nanobody Assembled via Ferritin Nanocage. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308248. [PMID: 38491904 PMCID: PMC11132087 DOI: 10.1002/advs.202308248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/12/2024] [Indexed: 03/18/2024]
Abstract
Increasing immunotherapy response rate and durability can lead to significant improvements in cancer care. To address this challenge, a novel multivalent immune checkpoint therapeutic platform is constructed through site-specific ligation of anti-PD-L1 nanobody (Nb) on ferritin (Ftn) nanocage. Nb-Ftn blocks PD-1/PD-L1 interaction and downregulates PD-L1 levels via endocytosis-induced degradation. In addition, the cage structure of Ftn allows encapsulation of indocyanine green (ICG), an FDA-approved dye. Photothermal treatment with Nb-Ftn@ICG induces immunogenic death of tumor cells, which improves systemic immune response via maturation of dendritic cells and enhanced infiltration of T cells. Moreover, Nb-Ftn encapsulation significantly enhances cellular uptake, tumor accumulation and retention of ICG. In vivo assays showed that this nanoplatform ablates the primary tumor, suppresses abscopal tumors and inhibits tumor metastasis, leading to a prolonged survival rate. This work presents a novel strategy for improving cancer immunotherapy using multivalent nanobody-ferritin conjugates as immunological targeting and enhancing carriers.
Collapse
Affiliation(s)
- Manman Liu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Duo Jin
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Wenxin Yu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Jiaji Yu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Kaiming Cao
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Junjie Cheng
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| | - Xiaohu Zheng
- The CAS Key Laboratory of Innate Immunity and Chronic DiseaseSchool of Basic Medical SciencesCenter for Advanced Interdisciplinary Science and Biomedicine of IHMDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230027China
| | - Andrew Wang
- Department of Radiation OncologyUniversity of Texas Southwestern Medical CenterDallas75230USA
| | - Yangzhong Liu
- Department of Pharmacythe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineDepartment of ChemistryUniversity of Science and Technology of ChinaHefei230001China
| |
Collapse
|
11
|
Wei J, Zheng H, Dai S, Liu M. A bibliometric and knowledge-map analysis of bispecific antibodies in cancer immunotherapy from 2000 to 2023. Heliyon 2024; 10:e23929. [PMID: 38312701 PMCID: PMC10835268 DOI: 10.1016/j.heliyon.2023.e23929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 02/06/2024] Open
Abstract
Background Bispecific antibody (BsAb)-based cancer immunotherapy has provided new avenues for the treatment of various malignancies. The approval of Blinatumomab has encouraged further investigation into these treatments, and a series of preclinical and clinical trials have been conducted, together with the publication of numerous articles. Here, the knowledge structure of BsAb-based cancer immunotherapy is summarized using bibliometric analysis to provide in-depth insight into current research trends and foci. Methods The studies included in the bibliometric analysis of BsAbs in cancer immunotherapy were retrieved from the online Web of Science Core Collection (WOSCC) database on April 16th, 2023. Visualization analysis was performed with the help of CtieSpace (version 6.2.2.msi [64-bit]), VOSviewer (version 1.6.19), R (version 4.2.1), and the Bibliometric analysis platform (R-based online data processing tool). Results A total of 1750 papers were identified. Analysis of annual publications and total citations indicated that publications have increased steadily over the past few decades. The USA, followed by Germany, had largest number of publications, making significant contributions to the field. The Memorial Sloan Kettering Cancer Center received the highest number of citations (n = 3769). However, its collaboration and cooperation with different institutions require further strengthening. MAbs and Clinical Cancer Research published the most papers, while Blood and Cancer Research were the most commonly co-cited journals. DM Goldenberg from the USA published the most articles with the highest H-index (34), and the most co-cited author (2137 citations) was PA Baeuerle; both these authors have distinguished achievements in this field. Analysis of co-cited references and keywords showed that the hotspots and research focus on the use of BsAbs for solid tumors have increased rapidly while the application of BsAb immunotherapy in hematologic malignancies has expanded significantly. The hot topics in the field included cytokine release syndrome, the efficacy and safety of BsAbs, resistance mechanisms, and the exploration and optimization of combination therapies. Conclusion Cancer immunotherapies based on BsAbs are a hot topic in research. Current studies focus on the construction and optimization of BsAb structure, as well as their combination with other treatment modalities to improve their efficacy and overcome resistance. Furthermore, it is expected that the ongoing investigation of BsAb-based immunotherapy for solid tumors will bear fruit with significant clinical application prospects in the near future.
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Huilan Zheng
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, 610075, China
| | - Shuang Dai
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ming Liu
- Department of Medical Oncology/Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| |
Collapse
|
12
|
Li L, Chen G, Chen EY, Strickland MR, Zhao W, Zhang J, Li Z. Development and validation of a nomogram to predict pathological complete response in patients with locally advanced gastric adenocarcinoma treated with neoadjuvant chemotherapy in combination with PD-1 antibodies. J Gastrointest Oncol 2023; 14:2373-2383. [PMID: 38196541 PMCID: PMC10772673 DOI: 10.21037/jgo-23-751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/20/2023] [Indexed: 01/11/2024] Open
Abstract
Background Currently, the survival benefits of combining neoadjuvant chemotherapy with programmed death 1 (PD-1) antibody immunotherapy in advanced gastric adenocarcinoma remain controversial. Emerging evidence suggests that the survival benefits of neoadjuvant therapy in advanced gastric adenocarcinoma hinge upon the attainment of pathological complete response (pCR). Therefore, the prediction of pCR in patients undergoing neoadjuvant chemotherapy combined with PD-1 antibody immunotherapy holds significant importance and is beneficial for the individualized treatment of gastric cancer (GC) patients. Methods Clinical and pathological characteristics of patients with GC who received neoadjuvant chemotherapy combined with PD-1 inhibitor (camrelizumab) therapy and radical gastrectomy between January 2019 and December 2020 at the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital were retrospectively analyzed. A total of 52 patients were enrolled in the study, with all subjects assigned to the training set. The neoadjuvant regimen consisted of a combination of PD-1 inhibitor and fluorouracil analogues plus oxaliplatin, comprising two drugs. The patients were divided into a pCR group and a non-pCR group according to pCR occurrence. Multifactor logistic regression analysis was applied to determine the correlation between each factor and pCR. A prediction model was developed based on the results of the logistic regression analysis. The predictive performance of the model was evaluated using the receiver operating characteristic curves. Internal verification was completed via the bootstrapping method. Results The pCR was observed in 10 out of 52 patients (19.2%). The results of binary logistic regression multivariate analysis showed that cN stage [odds ratio (OR): 0.215; P=0.03], combined positive score (CPS) (OR: 6.364; P=0.026), and tumor diameter (OR: 0.112; P=0.026) were independent predictors of pCR. The nomogram prediction model for the pCR was plotted with a concordance index of 0.923 [95% confidence interval (CI): 0.8441-1]. Conclusions Neoadjuvant chemotherapy combined with PD-1 antibodies may be the preferred option for patients with advanced gastric adenocarcinoma who have a small tumor diameter, no or few lymph node metastases, and high CPS. The presented nomogram model exhibits the potential to predict pCR in advanced gastric adenocarcinoma patients, showcasing satisfactory predictive performance and potentially facilitating the implementation of personalized treatment strategies.
Collapse
Affiliation(s)
- Liang Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Guanglong Chen
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Emerson Y. Chen
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Matthew R. Strickland
- Department of Medicine, Harvard Medical School and Massachusetts General Hospital, Boston, MA, USA
| | - Weijie Zhao
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Jialin Zhang
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Zhi Li
- Department of General Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
13
|
Bogut A, Stojanovic B, Jovanovic M, Dimitrijevic Stojanovic M, Gajovic N, Stojanovic BS, Balovic G, Jovanovic M, Lazovic A, Mirovic M, Jurisevic M, Jovanovic I, Mladenovic V. Galectin-1 in Pancreatic Ductal Adenocarcinoma: Bridging Tumor Biology, Immune Evasion, and Therapeutic Opportunities. Int J Mol Sci 2023; 24:15500. [PMID: 37958483 PMCID: PMC10650903 DOI: 10.3390/ijms242115500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most challenging malignancies to treat, with a complex interplay of molecular pathways contributing to its aggressive nature. Galectin-1 (Gal-1), a member of the galectin family, has emerged as a pivotal player in the PDAC microenvironment, influencing various aspects from tumor growth and angiogenesis to immune modulation. This review provides a comprehensive overview of the multifaceted role of Galectin-1 in PDAC. We delve into its contributions to tumor stroma remodeling, angiogenesis, metabolic reprogramming, and potential implications for therapeutic interventions. The challenges associated with targeting Gal-1 are discussed, given its pleiotropic functions and complexities in different cellular conditions. Additionally, the promising prospects of Gal-1 inhibition, including the utilization of nanotechnology and theranostics, are highlighted. By integrating recent findings and shedding light on the intricacies of Gal-1's involvement in PDAC, this review aims to provide insights that could guide future research and therapeutic strategies.
Collapse
Affiliation(s)
- Ana Bogut
- City Medical Emergency Department, 11000 Belgrade, Serbia;
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.); (G.B.)
- Department of General Surgery, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia;
| | - Marina Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.J.); (V.M.)
| | | | - Nevena Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Bojana S. Stojanovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Goran Balovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.); (G.B.)
| | - Milan Jovanovic
- Department of Abdominal Surgery, Military Medical Academy, 11000 Belgrade, Serbia;
| | - Aleksandar Lazovic
- Department of General Surgery, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia;
| | - Milos Mirovic
- Department of Surgery, General Hospital of Kotor, 85330 Kotor, Montenegro;
| | - Milena Jurisevic
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Violeta Mladenovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.J.); (V.M.)
| |
Collapse
|
14
|
Muacevic A, Adler JR, Alharbi R, Alrubaiaan A, Abdel-Razaq W, Alyousif G, Alkaiyat M. The Impact of Fasting the Holy Month of Ramadan on Colorectal Cancer Patients and Two Tumor Biomarkers: A Tertiary-Care Hospital Experience. Cureus 2023; 15:e33920. [PMID: 36819321 PMCID: PMC9936918 DOI: 10.7759/cureus.33920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Fasting during the holy month of Ramadan is a religious ritual practiced by the majority of Muslims around the globe. This daytime fasting is short-term or intermittent fasting, which may be associated with valuable health benefits, particularly in cancer patients. METHODS A prospective cohort study of pre- and post-fasting evaluation of 37 colorectal cancer (CRC) patients was conducted at King Abdulaziz Medical City (KAMC) and King Abdullah Specialized Children's Hospital (KASCH)-oncology outpatient clinics. The study aimed to assess the impact of fasting during the holy month of Ramadan on the tolerability of chemotherapy side effects and to assess changes in the levels of carcinoembryonic antigen (CEA) and lactate dehydrogenase (LDH) tumor biomarkers, which are primarily associated with certain types of carcinomas, including CRC. RESULTS A total of 33 patients (89.2%) had fasted at least part of the month of Ramadan. Twenty-seven patients (73%) reported "Serenity" after fasting during Ramadan with improved tolerability of chemotherapy side effects. However, the results did not reveal any significant difference in the measured laboratory variables between pre-fasting values and by the end of the 30 days of Ramadan. Although statistically insignificant, the levels of CEA and LDH were reduced in 46.9% and 55.6% of patients, respectively. The mean level of CEA in the fasting group was substantially reduced by more than 40%, attributed to the highly significant decline of CEA levels in three patients (p=0.0283). Moreover, there were no significant differences between pre- and post-fasting blood creatinine levels or estimated glomerular filtration rates, ruling out any possible adverse effects of fasting on renal function. CONCLUSION The current study confirms the safety and tolerability of intermittent fasting in CRC patients actively receiving chemotherapy, which is consistent with several reports. Nonetheless, the results did not reveal a significant decrease in CEA and LDH tumor biomarkers.
Collapse
|
15
|
Li Z, Chu Z, Yang J, Qian H, Xu J, Chen B, Tian T, Chen H, Xu Y, Wang F. Immunogenic Cell Death Augmented by Manganese Zinc Sulfide Nanoparticles for Metastatic Melanoma Immunotherapy. ACS NANO 2022; 16:15471-15483. [PMID: 35981098 DOI: 10.1021/acsnano.2c08013] [Citation(s) in RCA: 113] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Both T-cell deprivation and insufficient tumor immunogenicity seriously hinder the efficacy of immune-mediated tumor destruction in melanoma. In this work, an amphiphilic polyethylene glycol-poly(2-hexoxy-2-oxo-1,3,2-dioxaphospholane) copolymer with a thermally sensitive flowable core (mPEG-b-PHEP) was chosen to incorporate IR780 dye and manganese zinc sulfide nanoparticles (ZMS) to form polymer micelles (denoted PPIR780-ZMS), which precisely controlled the release of ZMS after being triggered by near-infrared light (NIR). Mn2+-mediated chemodynamic therapy (CDT) by photothermal trigger boosted the generation of reactive oxygen species (ROS), making the PPIR780-ZMS smart bomblets in vivo. It was demonstrated that PPIR780-ZMS could maximize immunogenic cell death (ICD) in cancer, which is characterized by abundant damage-associated molecular pattern (DAMP) exposure. As a result, the cytotoxic T cells (CD8+) and helper T cells (CD4+) expanded and infiltrated the neoplastic foci, which further reprogrammed the suppressive tumor microenvironment (TME) against the primary tumor and pulmonary metastases with safe systemic cytokine expression. In addition, Mn2+-mediated cGAS-STING signaling pathway activation enhanced the antitumor immunity of this nanocomposite, providing a practical strategy for expanding the use of Mn-based nanostructures.
Collapse
Affiliation(s)
- Zhu Li
- Department of Dermatology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P. R. China
| | - Zhaoyou Chu
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| | - Juan Yang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| | - Jiangmei Xu
- Department of Dermatology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P. R. China
| | - Benjin Chen
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| | - Tian Tian
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230036, P. R. China
| | - Hao Chen
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, Anhui 230032, P. R. China
| | - Yunsheng Xu
- Department of Dermatology, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P. R. China
| | - Fei Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong 518107, P. R. China
| |
Collapse
|
16
|
Wang N, Yang Y, Jin D, Zhang Z, Shen K, Yang J, Chen H, Zhao X, Yang L, Lu H. PARP inhibitor resistance in breast and gynecological cancer: Resistance mechanisms and combination therapy strategies. Front Pharmacol 2022; 13:967633. [PMID: 36091750 PMCID: PMC9455597 DOI: 10.3389/fphar.2022.967633] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/04/2022] [Indexed: 12/02/2022] Open
Abstract
Breast cancer and gynecological tumors seriously endanger women’s physical and mental health, fertility, and quality of life. Due to standardized surgical treatment, chemotherapy, and radiotherapy, the prognosis and overall survival of cancer patients have improved compared to earlier, but the management of advanced disease still faces great challenges. Recently, poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) have been clinically approved for breast and gynecological cancer patients, significantly improving their quality of life, especially of patients with BRCA1/2 mutations. However, drug resistance faced by PARPi therapy has hindered its clinical promotion. Therefore, developing new drug strategies to resensitize cancers affecting women to PARPi therapy is the direction of our future research. Currently, the effects of PARPi in combination with other drugs to overcome drug resistance are being studied. In this article, we review the mechanisms of PARPi resistance and summarize the current combination of clinical trials that can improve its resistance, with a view to identify the best clinical treatment to save the lives of patients.
Collapse
Affiliation(s)
- Nannan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Dongdong Jin
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China
| | - Zhenan Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ke Shen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huanhuan Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinyue Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China
- *Correspondence: Li Yang, ; Huaiwu Lu,
| | - Huaiwu Lu
- Department of Gynaecological Oncology, Sun Yat Sen Memorial Hospital, Guangzhou, China
- *Correspondence: Li Yang, ; Huaiwu Lu,
| |
Collapse
|
17
|
Lin JX, Lin JP, Weng Y, Lv CB, Chen JH, Zhan CY, Li P, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, Zhou WX, Zhang XJ, Zheng CH, Cai LS, Ma YB, Huang CM. Radiographical Evaluation of Tumor Immunosuppressive Microenvironment and Treatment Outcomes in Gastric Cancer: A Retrospective, Multicohort Study. Ann Surg Oncol 2022; 29:5022-5033. [PMID: 35532827 DOI: 10.1245/s10434-022-11499-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 02/05/2022] [Indexed: 11/18/2022]
|
18
|
Qin X, Zhang M, Zhao Z, Du Q, Li Q, Jiang Y, Xue F, Luan Y. A carrier-free photodynamic nanodrug to enable regulation of dendritic cells for boosting cancer immunotherapy. Acta Biomater 2022; 147:366-376. [PMID: 35588995 DOI: 10.1016/j.actbio.2022.05.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 11/01/2022]
Abstract
Immune response is initiated by dendritic cells (DCs), where the cross-presentation of antigens by DCs determines the activating of cytotoxic T cells. However, the efficacy of DCs-initiated immune response is governed by multiple (cascade) steps of immunogenic cell death (ICD), recruitment of DCs, and cross-presentation of DCs. It is urgent but challenging to achieve a platform for simultaneously regulating these multiple steps, amplifying the immune response against tumors. Herein, we reported a photodynamic nanodrug enabling simultaneous regulation of these multiple steps for realizing powerful immune response. The nanodrug was designed by the co-assembling of chlorin e6 (Ce6), celecoxib and 6-thio-2'-deoxyguanosine (6-thio-dG). In our nanodrug, Ce6 enables induction of ICD, while celecoxib down-regulates the prostaglandin E2 (PGE2) for promoting recruitment of DCs enabled by chemokine CCL5 produced from natural killer (NK) cells. Moreover, 6-thio-dG triggers DNA damages in the tumor cells, which in turn activates STING/interferon I pathway for enhancing the cross-presentation ability of DCs. Therefore, an amplified immune therapeutic effect against tumors is achieved, thanks to the simultaneous regulation of these multiple steps. The nanodrug effectively inhibits tumor growth and postoperative recurrence, demonstrating a new approach for boosting immune response initiated by DCs in cancer therapy. STATEMENT OF SIGNIFICANCE: The dendritic cells (DCs)-initiated immune response against tumors is dominated by multiple (cascade) steps including the process of (I) immunogenic cell death (ICD), (II) recruitment of DCs, and (III) cross-presentation of antigens by DCs. Based on this, it is urgent to design a nanoplatform enabling simultaneous regulation of these multiple steps for achieving a potent therapeutic efficacy. A carrier-free photodynamic nanodrug, engineered by a co-assembling approach, was designed to regulate DCs for realizing a powerful DCs-initiated immune response against tumors, thanks to the simultaneous regulation of the above multiple steps. Our nanodrug demonstrated a boosted immune response against tumors, powerfully suppressing primary/abscopal tumor growth and postoperative recurrence, which offers a conceptually innovative strategy for amplifying immunity against tumors.
Collapse
|
19
|
Application of additively manufactured 3D scaffolds for bone cancer treatment: a review. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00182-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
AbstractBone cancer is a critical health problem on a global scale, and the associated huge clinical and economic burdens are still rising. Although many clinical approaches are currently used for bone cancer treatment, these methods usually affect the normal body functions and thus present significant limitations. Meanwhile, advanced materials and additive manufacturing have opened up promising avenues for the development of new strategies targeting both bone cancer treatment and post-treatment bone regeneration. This paper presents a comprehensive review of bone cancer and its current treatment methods, particularly focusing on a number of advanced strategies such as scaffolds based on advanced functional materials, drug-loaded scaffolds, and scaffolds for photothermal/magnetothermal therapy. Finally, the main research challenges and future perspectives are elaborated.
Collapse
|
20
|
Engineered nanomaterials for synergistic photo-immunotherapy. Biomaterials 2022; 282:121425. [DOI: 10.1016/j.biomaterials.2022.121425] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/19/2022] [Accepted: 02/17/2022] [Indexed: 02/07/2023]
|
21
|
Wang M, Li Y, Wang M, Liu K, Hoover AR, Li M, Towner RA, Mukherjee P, Zhou F, Qu J, Chen WR. Synergistic interventional photothermal therapy and immunotherapy using an iron oxide nanoplatform for the treatment of pancreatic cancer. Acta Biomater 2022; 138:453-462. [PMID: 34757232 PMCID: PMC10960566 DOI: 10.1016/j.actbio.2021.10.048] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PC) is the most lethal malignancy due to its high metastatic ability and poor drug permeability. Here, a synergized interventional photothermal-immunotherapy strategy was developed with imaging guidance and temperature monitoring by magnetic resonance imaging (MRI) technique, for the local treatment of metastatic PC. A tumor microenvironment (TME)-responsive nanoplatform was fabricated via coating of DSPE-PEG and indocyanine green (ICG) onto imiquimod (IMQ) loaded amorphous iron oxide nanoparticles (IONs). This unique nanoplatform, IMQ@IONs/ICG, served as a contrast agent for MRI, a drug delivery vehicle for IMQ and ICG, and a catalyst for TME modulation. The biodegradable IMQ@IONs/ICG was also non-toxic, and improved the penetration of the loaded drugs in PC to maximize thermal ablation of the tumor and minimize damage to the surrounding healthy tissue. For the treatment of aggressive, metastatic Panc02-H7 pancreatic tumors in mice, ION-assisted MRI was employed to guide the administration of interventional photothermal therapy (IPTT) and monitor the temperature distribution in target tumor and surrounding tissue during treatment. The local IPTT treatment induced in situ immunogenic cell death (ICD), and, in combination with released IMQ, triggered a strong antitumor immunity, leading to decreased metastases and increased CD8+ in spleen and tumors. With precise local treatment and monitoring, treated primary tumors were completely eradicated, mesentery metastases were dramatically reduced, and the survival time was significantly prolonged, without damage to normal tissue and systemic autoimmunity. Overall, this synergistic strategy represents a promising approach to treat PC with significant potential for clinical applications. STATEMENT OF SIGNIFICANCE: Pancreatic cancer (PC) is one of the most lethal malignancies because it is non-permeable to drugs and highly metastatic. In this study, we designed a tumor microenvironment-responsive amorphous iron oxide nanoplatform (ION) to co-deliver photothermal agent (ICG) and toll-like-receptor-7 agonist (IMQ). This biodegradable nanoplatform IMQ@IONs/ICG improved the penetration of the loaded drugs in pancreatic tumor. With MR imaging guidance and temperature monitoring, the precise interventional photothermal therapy on mouse Panc02-H7 orthotopic tumors releases tumor antigens to initiate tumor-special immune responses, amplified by the released IMQ. Our results demonstrate that IMQ@IONs/ICG overcomes the obstacle of drug delivery to pancreatic tumors, and when combined with photothermal therapy, induces a systemic antitumor immunity to control metastatic tumors.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Yong Li
- Interventional Therapy Department, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Miao Wang
- School of Biomedical Engineering, Hainan University, Haikou 570228, China
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Feifan Zhou
- School of Biomedical Engineering, Hainan University, Haikou 570228, China.
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China.
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|
22
|
Pinkert J, Boehm HH, Trautwein M, Doecke WD, Wessel F, Ge Y, Gutierrez EM, Carretero R, Freiberg C, Gritzan U, Luetke-Eversloh M, Golfier S, Von Ahsen O, Volpin V, Sorrentino A, Rathinasamy A, Xydia M, Lohmayer R, Sax J, Nur-Menevse A, Hussein A, Stamova S, Beckmann G, Glueck JM, Schoenfeld D, Weiske J, Zopf D, Offringa R, Kreft B, Beckhove P, Willuda J. T cell-mediated elimination of cancer cells by blocking CEACAM6–CEACAM1 interaction. Oncoimmunology 2021; 11:2008110. [PMID: 35141051 PMCID: PMC8820806 DOI: 10.1080/2162402x.2021.2008110] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Carcinoembryonic antigen-related cell adhesion molecule 6 (CEACAM6), a cell surface receptor, is expressed on normal epithelial tissue and highly expressed in cancers of high unmet medical need, such as non-small cell lung, pancreatic, and colorectal cancer. CEACAM receptors undergo homo- and heterophilic interactions thereby regulating normal tissue homeostasis and angiogenesis, and in cancer, tumor invasion and metastasis. CEACAM6 expression on malignant plasma cells inhibits antitumor activity of T cells, and we hypothesize a similar function on epithelial cancer cells. The interactions between CEACAM6 and its suggested partner CEACAM1 on T cells were studied. A humanized CEACAM6-blocking antibody, BAY 1834942, was developed and characterized for its immunomodulating effects in co-culture experiments with T cells and solid cancer cells and in comparison to antibodies targeting the immune checkpoints programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and T cell immunoglobulin mucin-3 (TIM-3). The immunosuppressive activity of CEACAM6 was mediated by binding to CEACAM1 expressed by activated tumor-specific T cells. BAY 1834942 increased cytokine secretion by T cells and T cell-mediated killing of cancer cells. The in vitro efficacy of BAY 1834942 correlated with the degree of CEACAM6 expression on cancer cells, suggesting potential in guiding patient selection. BAY 1834942 was equally or more efficacious compared to blockade of PD-L1, and at least an additive efficacy was observed in combination with anti-PD-1 or anti-TIM-3 antibodies, suggesting an efficacy independent of the PD-1/PD-L1 axis. In summary, CEACAM6 blockade by BAY 1834942 reactivates the antitumor response of T cells. This warrants clinical evaluation.
Collapse
Affiliation(s)
- Jessica Pinkert
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hans-Henning Boehm
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | | | - Florian Wessel
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Yingzi Ge
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Maria Gutierrez
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rafael Carretero
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | | - Uwe Gritzan
- Pharmaceutical Division, Bayer AG, Cologne, Germany
| | | | - Sven Golfier
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | | | - Valentina Volpin
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Antonio Sorrentino
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Anchana Rathinasamy
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Maria Xydia
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Robert Lohmayer
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
- Institute of Theoretical Physics, University of Regensburg, Regensburg, Germany
| | - Julian Sax
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Ayse Nur-Menevse
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Abir Hussein
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | - Slava Stamova
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
| | | | | | | | - Joerg Weiske
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | - Dieter Zopf
- Pharmaceutical Division, Bayer AG, Berlin, Germany
| | - Rienk Offringa
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Philipp Beckhove
- Joint Immunotherapeutics Laboratory of the DKFZ-Bayer Innovation Alliance, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Regensburg, Germany
- Hematology and Oncology Department, University Hospital Regensburg, Regensburg, Germany
| | | |
Collapse
|
23
|
Wan D, Que H, Chen L, Lan T, Hong W, He C, Yang J, Wei Y, Wei X. Lymph-Node-Targeted Cholesterolized TLR7 Agonist Liposomes Provoke a Safe and Durable Antitumor Response. NANO LETTERS 2021; 21:7960-7969. [PMID: 34533963 DOI: 10.1021/acs.nanolett.1c01968] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Toll-like receptor (TLR) agonists as the potent stimulants of an innate immune system hold promises for applications in anticancer immunotherapy. However, most of them are limited in the clinical translation due to the uncontrolled systemic inflammatory response. In the current study, 1V209, a small molecule TLR7 agonist, was conjugated with cholesterol (1V209-Cho) and prepared into liposomes (1V209-Cho-Lip). 1V209-Cho-Lip exerted minimal toxic effects and enhanced the transportation ability in lymph nodes (LNs) compared with 1V209. 1V209-Cho-Lip treatment inhibited tumor progression in CT26 colorectal cancer, 4T1 breast cancer, and Pan02 pancreatic ductal cancer models through inducing effective DC activation and eliciting CD8+ T cell responses. Furthermore, 1V209-Cho-Lip induced tumor-specific memory immunity to inhibit cancer recurrence and metastasis. These results indicate that cholesterol conjugation with 1V209 is an effective approach to target lymph nodes and to reduce the adverse effects. This work provides a rational basis for the distribution optimization of TLR agonists for potential clinical use.
Collapse
Affiliation(s)
- Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Haiying Que
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Li Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Cai He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
24
|
Tan CL, Kuchroo JR, Sage PT, Liang D, Francisco LM, Buck J, Thaker YR, Zhang Q, McArdel SL, Juneja VR, Lee SJ, Lovitch SB, Lian C, Murphy GF, Blazar BR, Vignali DAA, Freeman GJ, Sharpe AH. PD-1 restraint of regulatory T cell suppressive activity is critical for immune tolerance. J Exp Med 2021; 218:191205. [PMID: 33045061 PMCID: PMC7543091 DOI: 10.1084/jem.20182232] [Citation(s) in RCA: 172] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/30/2020] [Accepted: 08/26/2020] [Indexed: 12/11/2022] Open
Abstract
Inhibitory signals through the PD-1 pathway regulate T cell activation, T cell tolerance, and T cell exhaustion. Studies of PD-1 function have focused primarily on effector T cells. Far less is known about PD-1 function in regulatory T (T reg) cells. To study the role of PD-1 in T reg cells, we generated mice that selectively lack PD-1 in T reg cells. PD-1–deficient T reg cells exhibit an activated phenotype and enhanced immunosuppressive function. The in vivo significance of the potent suppressive capacity of PD-1–deficient T reg cells is illustrated by ameliorated experimental autoimmune encephalomyelitis (EAE) and protection from diabetes in nonobese diabetic (NOD) mice lacking PD-1 selectively in T reg cells. We identified reduced signaling through the PI3K–AKT pathway as a mechanism underlying the enhanced suppressive capacity of PD-1–deficient T reg cells. Our findings demonstrate that cell-intrinsic PD-1 restraint of T reg cells is a significant mechanism by which PD-1 inhibitory signals regulate T cell tolerance and autoimmunity.
Collapse
Affiliation(s)
- Catherine L Tan
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Juhi R Kuchroo
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Peter T Sage
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Dan Liang
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Loise M Francisco
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Jessica Buck
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Youg Raj Thaker
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA.,School of Life Sciences, University of Essex, Wivenhoe Park, Colchester, UK
| | - Qianxia Zhang
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Shannon L McArdel
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Vikram R Juneja
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Sun Jung Lee
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA
| | - Scott B Lovitch
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Christine Lian
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - George F Murphy
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota Medical School, Twin Cities, MN
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA.,Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA.,Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh PA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA.,Harvard Medical School, Boston, MA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA.,Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
25
|
Mensali N, Dillard P, Fayzullin A, Köksal H, Gaudernack G, Kvalheim G, Inderberg EM, Wälchli S. "Built-in" PD-1 blocker to rescue NK-92 activity from PD-L1-mediated tumor escape mechanisms. FASEB J 2021; 35:e21750. [PMID: 34424568 DOI: 10.1096/fj.202100025r] [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: 01/08/2021] [Revised: 05/11/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
Success of adoptive cell therapy mainly depends on the ability of immune cells to persist and function optimally in the immunosuppressive tumor microenvironment. Although present at the cancer site, immune cells become exhausted and/or inhibited, due to the presence of inhibitory receptors such as PD-L1 on malignant cells. Novel genetic strategies to manipulate the PD1/PD-L1 axis comprise (i) PD-1 reversion where the receptor intracellular domain is replaced with an activating unit, (ii) the use of anti-PD-L1 CAR or (iii) the disruption of the PD-1 gene. We here present an alternative strategy to equip therapeutic cells with a truncated PD-1 (tPD-1) to abrogate PD-1/PD-L1 inhibition. We show that engagement of tPD-1 with PD-L1-positive tumor unleashes NK-92 activity in vitro. Furthermore, this binding was sufficiently strong to induce killing of targets otherwise not recognized by NK-92, thus increasing the range of targets. In vivo treatment with NK-92 tPD-1 cells led to reduced tumor growth and improved survival. Importantly, tPD-1 did not interfere with tumor recognition in PD-L1 negative conditions. Thus, tPD-1 represents a straightforward method for improving antitumor immunity and revealing new targets through PD-L1 positivity.
Collapse
Affiliation(s)
- Nadia Mensali
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Pierre Dillard
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Artem Fayzullin
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Hakan Köksal
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Gustav Gaudernack
- Department of Cancer Immunology, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Department of Cellular Therapy, Oslo University Hospital-The Norwegian Radium Hospital, Oslo, Norway
| |
Collapse
|
26
|
Li CL, Song Y. Combination strategies of immunotherapy in non-small cell lung cancer: facts and challenges. Chin Med J (Engl) 2021; 134:1908-1919. [PMID: 34343148 PMCID: PMC8382384 DOI: 10.1097/cm9.0000000000001610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Indexed: 12/09/2022] Open
Abstract
ABSTRACT Immunotherapy has dramatically altered the treatment of non-small cell lung cancer. Currently, the emergence of combination strategies in immunotherapy has brightened the prospects of improved clinical outcomes and manageable safety profiles in the first/second-line settings. However, sub-optimal response rates are still observed in several clinical trials. Hence, alternative combination models and candidate selection strategies need to be explored. Herein, we have critically reviewed and commented on the published data from several clinical trials, including combined immunotherapy and chemotherapy, anti-angiogenic agents, epidermal growth factor receptor/anaplastic lymphoma kinase tyrosine kinase inhibitors, radiotherapy, and other immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Chu-Ling Li
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | | |
Collapse
|
27
|
Di Pompo G, Cortini M, Baldini N, Avnet S. Acid Microenvironment in Bone Sarcomas. Cancers (Basel) 2021; 13:cancers13153848. [PMID: 34359749 PMCID: PMC8345667 DOI: 10.3390/cancers13153848] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Although rare, malignant bone sarcomas have devastating clinical implications for the health and survival of young adults and children. To date, efforts to identify the molecular drivers and targets have focused on cancer cells or on the interplay between cancer cells and stromal cells in the tumour microenvironment. On the contrary, in the current literature, the role of the chemical-physical conditions of the tumour microenvironment that may be implicated in sarcoma aggressiveness and progression are poorly reported and discussed. Among these, extracellular acidosis is a well-recognized hallmark of bone sarcomas and promotes cancer growth and dissemination but data presented on this topic are fragmented. Hence, we intended to provide a general and comprehensive overview of the causes and implications of acidosis in bone sarcoma. Abstract In bone sarcomas, extracellular proton accumulation is an intrinsic driver of malignancy. Extracellular acidosis increases stemness, invasion, angiogenesis, metastasis, and resistance to therapy of cancer cells. It reprograms tumour-associated stroma into a protumour phenotype through the release of inflammatory cytokines. It affects bone homeostasis, as extracellular proton accumulation is perceived by acid-sensing ion channels located at the cell membrane of normal bone cells. In bone, acidosis results from the altered glycolytic metabolism of bone cancer cells and the resorption activity of tumour-induced osteoclasts that share the same ecosystem. Proton extrusion activity is mediated by extruders and transporters located at the cell membrane of normal and transformed cells, including vacuolar ATPase and carbonic anhydrase IX, or by the release of highly acidic lysosomes by exocytosis. To date, a number of investigations have focused on the effects of acidosis and its inhibition in bone sarcomas, including studies evaluating the use of photodynamic therapy. In this review, we will discuss the current status of all findings on extracellular acidosis in bone sarcomas, with a specific focus on the characteristics of the bone microenvironment and the acid-targeting therapeutic approaches that are currently being evaluated.
Collapse
Affiliation(s)
- Gemma Di Pompo
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Margherita Cortini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
| | - Nicola Baldini
- Biomedical Science and Technologies Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (G.D.P.); (M.C.); (N.B.)
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy
- Correspondence:
| |
Collapse
|
28
|
Chan DKH, Buczacki SJA. Tumour heterogeneity and evolutionary dynamics in colorectal cancer. Oncogenesis 2021; 10:53. [PMID: 34272358 PMCID: PMC8285471 DOI: 10.1038/s41389-021-00342-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/21/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Colorectal cancer (CRC) has a global burden of disease. Our current understanding of CRC has progressed from initial discoveries which focused on the stepwise accumulation of key driver mutations, as encapsulated in the Vogelstein model, to one in which marked heterogeneity leads to a complex interplay between clonal populations. Current evidence suggests that an initial explosion, or “Big Bang”, of genetic diversity is followed by a period of neutral dynamics. A thorough understanding of this interplay between clonal populations during neutral evolution gives insights into the roles in which driver genes may participate in the progress from normal colonic epithelium to adenoma and carcinoma. Recent advances have focused not only on genetics, transcriptomics, and proteomics but have also investigated the ecological and evolutionary processes which transform normal cells into cancer. This review first describes the role which driver mutations play in the Vogelstein model and subsequently demonstrates the evidence which supports a more complex model. This article also aims to underscore the significance of tumour heterogeneity and diverse clonal populations in cancer progression.
Collapse
Affiliation(s)
- Dedrick Kok Hong Chan
- Nuffield Department of Surgical Sciences, Medical Sciences Division, University of Oxford, Oxford, UK
| | | |
Collapse
|
29
|
Zhai T, Wang C, Xu Y, Huang W, Yuan Z, Wang T, Dai S, Peng S, Pang T, Jiang W, Huang Y, Zou Y, Xu Y, Sun J, Gong X, Zhang J, Tsun A, Li B, Miao X. Generation of a safe and efficacious llama single-domain antibody fragment (vHH) targeting the membrane-proximal region of 4-1BB for engineering therapeutic bispecific antibodies for cancer. J Immunother Cancer 2021; 9:jitc-2020-002131. [PMID: 34172514 PMCID: PMC8237747 DOI: 10.1136/jitc-2020-002131] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2021] [Indexed: 12/16/2022] Open
Abstract
Background The discovery of checkpoint inhibitors towards cytotoxic T-lymphocyte protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) has been revolutionary for the treatment of cancers. These therapies have only offered an average of 20%–30% response rates across the tumor spectrum and the combination of agonists towards the tumor-necrosis superfamily members, such as 4-1BB and CD40, has shown potent efficacy in preclinical studies; however, these agonists have exhibited high degrees of toxicity with limited efficacy in human trials. In this study, we have generated a single-domain antibody towards a unique epitope of 4-1BB that limits its potential on-target toxicity while maintaining sufficient potency. This 4-1BB binder is ideal for use in the engineering of multispecific antibodies to localize 4-1BB activation within the tumor microenvironment, as shown here by a anti-PD-L1/4-1BB bispecific candidate (PM1003). Methods To determine the functional activity of the 4-1BB- and PD-L1-binding elements of PM1003, in vitro luciferase reporter and primary cell assays were used to test the potency of programmed cell death 1 ligand 1 (PD-L1) blockade and PD-L1-mediated 4-1BB activation via cross-bridging. X-ray crystallography was conducted to resolve the binding epitopes of the respective binding arms, and accurate binding kinetics were determined using standard affinity measurement techniques. Human 4-1BB and/or PD-L1 knock-in mice were used in cancer models for testing the in vivo antitumor efficacy of PM1003, and safety was evaluated further. Results PM1003 shows potent activation of 4-1BB and blockade of PD-L1 in cell-based assays. 4-1BB activation was exerted through the bridging of PD-L1 on target cells and 4-1BB on effector cells. No PD-L1-independent activation of 4-1BB was observed. Through X-ray crystallography, a unique binding epitope in the cysteine-rich domain 4 (CRD4) region was resolved that provides high potency and potentially low on-target toxicity as determined by primary immune cell assays and toxicity evaluation in vivo. Conclusions A unique single-domain antibody was discovered that binds to the CRD4 domain of 4-1BB. When incorporated into a 4-1BB/PD-L1 bispecific (PM1003), we have shown the potent inhibition of PD-L1 activity with 4-1BB agonism upon cross-bridging with PD-L1 in vitro. Antitumor activity with minimal toxicity was found in vivo. Thus, PM1003 is a uniquely differentiating and next generation therapeutic agent for cancer therapy.
Collapse
Affiliation(s)
- Tianhang Zhai
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Wang
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Yifeng Xu
- Discovery Biology, Biotheus (Suzhou) Co., Ltd, Suzhou, China
| | - Weifeng Huang
- Discovery Biology, Biotheus (Suzhou) Co., Ltd, Suzhou, China
| | - Zhijun Yuan
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Tao Wang
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Shuang Dai
- Discovery Biology, Biotheus (Suzhou) Co., Ltd, Suzhou, China
| | - Shaogang Peng
- Discovery Biology, Biotheus (Suzhou) Co., Ltd, Suzhou, China
| | - Tuling Pang
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Wenchao Jiang
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Yuhua Huang
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Yuefeng Zou
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Yingda Xu
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Joanne Sun
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Xinjiang Gong
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Andy Tsun
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoniu Miao
- Discovery Biology & Discovery Technology, Biotheus Inc, Zhuhai, China .,Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
30
|
Yoon JH, Jung YJ, Moon SH. Immunotherapy for pancreatic cancer. World J Clin Cases 2021; 9:2969-2982. [PMID: 33969083 PMCID: PMC8080736 DOI: 10.12998/wjcc.v9.i13.2969] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/03/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer, a highly lethal cancer, has the lowest 5-year survival rate for several reasons, including its tendency for the late diagnosis, a lack of serologic markers for screening, aggressive local invasion, its early metastatic dissemination, and its resistance to chemotherapy/radiotherapy. Pancreatic cancer evades immunologic elimination by a variety of mechanisms, including induction of an immunosuppressive microenvironment. Cancer-associated fibroblasts interact with inhibitory immune cells, such as tumor-associated macrophages and regulatory T cells, to form an inflammatory shell-like desmoplastic stroma around tumor cells. Immunotherapy has the potential to mobilize the immune system to eliminate cancer cells. Nevertheless, although immunotherapy has shown brilliant results across a wide range of malignancies, only anti-programmed cell death 1 antibodies have been approved for use in patients with pancreatic cancer who test positive for microsatellite instability or mismatch repair deficiency. Some patients treated with immunotherapy who show progression based on conventional response criteria may prove to have a durable response later. Continuation of immune-based treatment beyond disease progression can be chosen if the patient is clinically stable. Immunotherapeutic approaches for pancreatic cancer treatment deserve further exploration, given the plethora of combination trials with other immunotherapeutic agents, targeted therapy, stroma-modulating agents, chemotherapy, and multi-way combination therapies.
Collapse
Affiliation(s)
- Jai Hoon Yoon
- Department of Internal Medicine, Hanyang University College of Medicine, Seoul 04763, South Korea
| | - Ye-Ji Jung
- Department of Internal Medicine, Hallym University, Anyang 14068, South Korea
| | - Sung-Hoon Moon
- Department of Internal Medicine, University of Hallym College of Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, South Korea
| |
Collapse
|
31
|
Investigating T Cell Immunity in Cancer: Achievements and Prospects. Int J Mol Sci 2021; 22:ijms22062907. [PMID: 33809369 PMCID: PMC7999898 DOI: 10.3390/ijms22062907] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/04/2021] [Accepted: 03/10/2021] [Indexed: 12/21/2022] Open
Abstract
T cells play a key role in tumour surveillance, both identifying and eliminating transformed cells. However, as tumours become established they form their own suppressive microenvironments capable of shutting down T cell function, and allowing tumours to persist and grow. To further understand the tumour microenvironment, including the interplay between different immune cells and their role in anti-tumour immune responses, a number of studies from mouse models to clinical trials have been performed. In this review, we examine mechanisms utilized by tumour cells to reduce their visibility to CD8+ Cytotoxic T lymphocytes (CTL), as well as therapeutic strategies trialled to overcome these tumour-evasion mechanisms. Next, we summarize recent advances in approaches to enhance CAR T cell activity and persistence over the past 10 years, including bispecific CAR T cell design and early evidence of efficacy. Lastly, we examine mechanisms of T cell infiltration and tumour regression, and discuss the strengths and weaknesses of different strategies to investigate T cell function in murine tumour models.
Collapse
|
32
|
Reina-Ortiz C, Constantinides M, Fayd-Herbe-de-Maudave A, Présumey J, Hernandez J, Cartron G, Giraldos D, Díez R, Izquierdo I, Azaceta G, Palomera L, Marzo I, Naval J, Anel A, Villalba M. Expanded NK cells from umbilical cord blood and adult peripheral blood combined with daratumumab are effective against tumor cells from multiple myeloma patients. Oncoimmunology 2020; 10:1853314. [PMID: 33457074 PMCID: PMC7781838 DOI: 10.1080/2162402x.2020.1853314] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In this study we evaluated the potential of expanded NK cells (eNKs) from two sources combined with the mAbs daratumumab and pembrolizumab to target primary multiple myeloma (MM) cells ex vivo. In order to ascertain the best source of NK cells, we expanded and activated NK cells from peripheral blood (PB) of healthy adult donors and from umbilical cord blood (UCB). The resulting expanded NK (eNK) cells express CD16, necessary for carrying out antibody-dependent cellular cytotoxicity (ADCC). Cytotoxicity assays were performed on bone marrow aspirates of 18 MM patients and 4 patients with monoclonal gammopathy of undetermined significance (MGUS). Expression levels of PD-1 on eNKs and PD-L1 on MM and MGUS cells were also quantified. Results indicate that most eNKs obtained using our expansion protocol express a low percentage of PD-1+ cells. UCB eNKs were highly cytotoxic against MM cells and addition of daratumumab or pembrolizumab did not further increase their cytotoxicity. PB eNKs, while effective against MM cells, were significantly more cytotoxic when combined with daratumumab. In a minority of cases, eNK cells showed a detectable population of PD1+ cells. This correlated with low cytotoxic activity, particularly in UCB eNKs. Addition of pembrolizumab did not restore their activity. Results indicate that UCB eNKs are to be preferentially used against MM in the absence of daratumumab while PB eNKs have significant cytotoxic advantage when combined with this mAb.
Collapse
Affiliation(s)
- Chantal Reina-Ortiz
- Apoptosis, Immunity & Cancer Group, Dept. Biochemistry and Molecular and Cell Biology, Faculty of Sciences, Campus San Francisco Sq., University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | | | | | | | | | | | - David Giraldos
- Apoptosis, Immunity & Cancer Group, Dept. Biochemistry and Molecular and Cell Biology, Faculty of Sciences, Campus San Francisco Sq., University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Rosana Díez
- Hematology Department, Miguel Servet Hospital, Zaragoza, Spain
| | | | - Gemma Azaceta
- Hematology Department, Lozano Blesa Hospital, Zaragoza, Spain
| | - Luis Palomera
- Hematology Department, Lozano Blesa Hospital, Zaragoza, Spain
| | - Isabel Marzo
- Apoptosis, Immunity & Cancer Group, Dept. Biochemistry and Molecular and Cell Biology, Faculty of Sciences, Campus San Francisco Sq., University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Javier Naval
- Apoptosis, Immunity & Cancer Group, Dept. Biochemistry and Molecular and Cell Biology, Faculty of Sciences, Campus San Francisco Sq., University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Alberto Anel
- Apoptosis, Immunity & Cancer Group, Dept. Biochemistry and Molecular and Cell Biology, Faculty of Sciences, Campus San Francisco Sq., University of Zaragoza and Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Martín Villalba
- CHU Montpellier, IRMB, Montpellier, France.,IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| |
Collapse
|
33
|
Chen H, Song Y, Deng C, Xu Y, Xu H, Zhu X, Song G, Tang Q, Lu J, Wang J. Comprehensive analysis of immune infiltration and gene expression for predicting survival in patients with sarcomas. Aging (Albany NY) 2020; 13:2168-2183. [PMID: 33316779 PMCID: PMC7880383 DOI: 10.18632/aging.202229] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/31/2020] [Indexed: 12/24/2022]
Abstract
Tumor microenvironments are strongly related to tumor development, and immune-infiltrating cells and immune-related molecules are potential prognostic markers. However, the shortcomings of traditional measurement methods limit the accurate evaluation of various components in tumor microenvironments. With the rapid advancement of Next-Generation RNA Sequencing technology, dedicated and in-depth analyses of immune filtration within the tumor microenvironment has been achieved. In this study, we combined the bioinformatics analysis methods ESTIMATE, CIBERSORT, and ssGSEA to characterize the immune infiltration of sarcomas and to identify specific immunomodulators of different pathological subtypes. We further extracted a functional enrichment of significant immune-related genes related to improved prognosis, including NR1H3, VAMP5, GIMAP2, GBP2, HLA-E and CRIP1. Overall, the immune microenvironment is an important prognostic determinant of sarcomas and may be a potential resource for developing effective immunotherapy.
Collapse
Affiliation(s)
- Hongmin Chen
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Yijiang Song
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Chuangzhong Deng
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Yanyang Xu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Huaiyuan Xu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Xiaojun Zhu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Guohui Song
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Qinglian Tang
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Jinchang Lu
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| | - Jin Wang
- Department of Musculoskeletal Oncology, Sun Yat-Sen University Cancer Center, Guangzhou 510060, Guangdong, P. R. China
- State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center of Cancer Medicine, Guangzhou 510060, P. R. China
| |
Collapse
|
34
|
Mielgo-Rubio X, Rojo F, Mezquita-Pérez L, Casas F, Wals A, Juan M, Aguado C, Garde-Noguera J, Vicente D, Couñago F. Deep diving in the PACIFIC: Practical issues in stage III non-small cell lung cancer to avoid shipwreck. World J Clin Oncol 2020; 11:898-917. [PMID: 33312885 PMCID: PMC7701908 DOI: 10.5306/wjco.v11.i11.898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/30/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
After publication of the PACIFIC trial results, immune checkpoint inhibitor-based immunotherapy was included in the treatment algorithm of locally advanced non-small cell lung cancer (NSCLC). The PACIFIC trial demonstrated that 12 mo of durvalumab consolidation therapy after radical-intent platinum doublet chemotherapy with concomitant radiotherapy improved both progression-free survival and overall survival in patients with unresectable stage III NSCLC. This is the first treatment in decades to successfully improve survival in this clinical setting, with manageable toxicity and without deterioration in quality of life. The integration of durvalumab in the management of locally advanced NSCLC accentuates the need for multidisciplinary, coordinated decision-making among lung cancer specialists, bringing new challenges and controversies as well as important changes in clinical work routines. The aim of the present article is to review-from a practical, multidisciplinary perspective-the findings and implications of the PACIFIC trial. We evaluate the immunobiological basis of durvalumab as well as practical aspects related to programmed cell death ligand 1 determination. In addition, we comprehensively assess the efficacy and toxicity data from the PACIFIC trial and discuss the controversies and practical aspects of incorporating durvalumab into routine clinical practice. Finally, we discuss unresolved questions and future challenges. In short, the present document aims to provide clinicians with a practical guide for the application of the PACIFIC regimen in routine clinical practice.
Collapse
Affiliation(s)
- Xabier Mielgo-Rubio
- Department of Medical Oncology, Hospital Universitario Fundación Alcorcón, Madrid 28922, Spain
| | - Federico Rojo
- Department of Pathology, IIS-Jiménez Díaz-CIBERONC Foundation, Madrid 28040, Spain
| | - Laura Mezquita-Pérez
- Department of Medical Oncology, Hospital Clinic, Laboratory of Translational Genomics and Targeted Therapeutics in Solid Tumors, IDIBAPS, Barcelona 08036, Spain
| | - Francesc Casas
- Department of Radiation Oncology, Hospital Clinic, Barcelona 08036, Spain
| | - Amadeo Wals
- Department of Radiation Oncology, Hospital Universitario Virgen Macarena, Sevilla 41009, Spain
| | - Manel Juan
- Department of Immunology Service, Hospital Clínic, Universitat de Barcelona, Barcelona 08036, Spain
| | - Carlos Aguado
- Department of Medical Oncology, Hospital Universitario Clínico San Carlos, Madrid 28040, Spain
| | - Javier Garde-Noguera
- Department of Medical Oncology, Hospital Arnau de Vilanova, Valencia 46015, Spain
| | - David Vicente
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Sevilla 49001, Spain
| | - Felipe Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud Madrid, Hospital La Luz, Universidad Europea de Madrid, Madrid 28028, Spain
| |
Collapse
|
35
|
Priem B, van Leent MMT, Teunissen AJP, Sofias AM, Mourits VP, Willemsen L, Klein ED, Oosterwijk RS, Meerwaldt AE, Munitz J, Prévot G, Vera Verschuur A, Nauta SA, van Leeuwen EM, Fisher EL, de Jong KAM, Zhao Y, Toner YC, Soultanidis G, Calcagno C, Bomans PHH, Friedrich H, Sommerdijk N, Reiner T, Duivenvoorden R, Zupančič E, Di Martino JS, Kluza E, Rashidian M, Ploegh HL, Dijkhuizen RM, Hak S, Pérez-Medina C, Bravo-Cordero JJ, de Winther MPJ, Joosten LAB, van Elsas A, Fayad ZA, Rialdi A, Torre D, Guccione E, Ochando J, Netea MG, Griffioen AW, Mulder WJM. Trained Immunity-Promoting Nanobiologic Therapy Suppresses Tumor Growth and Potentiates Checkpoint Inhibition. Cell 2020; 183:786-801.e19. [PMID: 33125893 PMCID: PMC8074872 DOI: 10.1016/j.cell.2020.09.059] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 03/25/2020] [Accepted: 09/23/2020] [Indexed: 12/17/2022]
Abstract
Trained immunity, a functional state of myeloid cells, has been proposed as a compelling immune-oncological target. Its efficient induction requires direct engagement of myeloid progenitors in the bone marrow. For this purpose, we developed a bone marrow-avid nanobiologic platform designed specifically to induce trained immunity. We established the potent anti-tumor capabilities of our lead candidate MTP10-HDL in a B16F10 mouse melanoma model. These anti-tumor effects result from trained immunity-induced myelopoiesis caused by epigenetic rewiring of multipotent progenitors in the bone marrow, which overcomes the immunosuppressive tumor microenvironment. Furthermore, MTP10-HDL nanotherapy potentiates checkpoint inhibition in this melanoma model refractory to anti-PD-1 and anti-CTLA-4 therapy. Finally, we determined MTP10-HDL's favorable biodistribution and safety profile in non-human primates. In conclusion, we show that rationally designed nanobiologics can promote trained immunity and elicit a durable anti-tumor response either as a monotherapy or in combination with checkpoint inhibitor drugs.
Collapse
Affiliation(s)
- Bram Priem
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands; Department of Medical Oncology (NA Angiogenesis Laboratory), Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Mandy M T van Leent
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandros Marios Sofias
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Vera P Mourits
- Department of Internal Medicine and Radboud Center of Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lisa Willemsen
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | - Emma D Klein
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Roderick S Oosterwijk
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anu E Meerwaldt
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Jazz Munitz
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Geoffrey Prévot
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anna Vera Verschuur
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sheqouia A Nauta
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Esther M van Leeuwen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Elizabeth L Fisher
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karen A M de Jong
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yiming Zhao
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yohana C Toner
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Soultanidis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul H H Bomans
- Department of Chemical Engineering and Chemistry, Center for Multiscale Electron Microscopy and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Heiner Friedrich
- Department of Chemical Engineering and Chemistry, Center for Multiscale Electron Microscopy and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Nico Sommerdijk
- Department of Biochemistry, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Thomas Reiner
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Raphaël Duivenvoorden
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Eva Zupančič
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie S Di Martino
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Ewelina Kluza
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Hidde L Ploegh
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Rick M Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Sjoerd Hak
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Carlos Pérez-Medina
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Jose Javier Bravo-Cordero
- Department of Medicine, Division of Hematology and Medical Oncology, Icahn School of Medicine, Tisch Cancer Institute at Mount Sinai, New York, NY, USA
| | - Menno P J de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Amsterdam Cardiovascular Sciences, Amsterdam Infection and Immunity, Amsterdam UMC, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Munich, Germany
| | - Leo A B Joosten
- Department of Internal Medicine and Radboud Institute of Molecular Life Sciences (RIMLS), Radboud University Medical Center, Nijmegen, the Netherlands; Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | | | - Zahi A Fayad
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Rialdi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denis Torre
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ernesto Guccione
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmacological Sciences and Mount Sinai Center for Therapeutics Discovery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center of Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania; Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Arjan W Griffioen
- Department of Medical Oncology (NA Angiogenesis Laboratory), Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, the Netherlands
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Laboratory of Chemical Biology, Department of Biochemical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
36
|
Liu S, Wang F, Tan W, Zhang L, Dai F, Wang Y, Fan Y, Yuan M, Yang D, Zheng Y, Deng Z, Liu Y, Cheng Y. CTLA4 has a profound impact on the landscape of tumor-infiltrating lymphocytes with a high prognosis value in clear cell renal cell carcinoma (ccRCC). Cancer Cell Int 2020; 20:519. [PMID: 33117084 PMCID: PMC7590466 DOI: 10.1186/s12935-020-01603-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Cytotoxic T-lymphocyte associated protein 4 (CTLA4) inhibitors have been shown to significantly prolong the overall survival (OS) in a wide range of cancers. However, its application in clear cell renal cell carcinoma (ccRCC) is limited due to the therapy response, and the prognostic value of CTLA4 in ccRCC has not been investigated in detail. Methods By using immunohistochemistry, Kaplan–Meier (K–M) analysis, uni- and multi-variate Cox analysis, we comprehensively and systematically studied the prognostic value of CTLA4 in ccRCC. Then, we applied Gene Ontology (GO), the Kyoto Encyclopedia of Genes and Genomes (KEGG) and CIBERSORT, ESTIMATE algorithm, ssGSEA and somatic mutation analyses to reveal the impact of CTLA4 on the landscape of tumor-infiltrating lymphocytes (TILs) infiltration and genetic mutation. Besides, given current concerns caused by combined immunotherapy, we also investigated the relationship between CTLA4 and other immune checkpoints. Results In vitro experiment and data mining showed that, CTLA4 was up-regulated in ccRCC tissues and closely related to the disease progression as well as a poor prognosis. Deeper researches demonstrated that CTLA4 regulates T cell activation and was significantly linked to TIL-abundant tumor microenvironment (TME), but was accompanied by an immunosuppressed phenotype. Mutation analysis showed that CTLA4 was associated with more frequent BRCA-associated protein 1 (BAP1) mutation. Moreover, we found that CTLA4 was markedly correlated with multiple immune checkpoints, which suggested that ccRCC patients with high expressed CTLA4 may benefit more from immune checkpoint blockades (ICBs) combined therapy. Conclusion CTLA4 has a profound impact on the landscape of TILs and genetic mutation, and can be used as the biomarker with high prognosis value in ccRCC.
Collapse
Affiliation(s)
- Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Feiyan Wang
- Shanghai Skin Disease Clinical College of Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200443 People's Republic of China
| | - Wei Tan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Li Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Yanqing Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Yaqi Fan
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200071 Shanghai People's Republic of China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Zhimin Deng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| | - Yeqiang Liu
- Department of Dermatopathology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, 200071 Shanghai People's Republic of China.,Department of Dermatopathology, Shanghai Skin Disease Clinical College of Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai, 200071 Shanghai People's Republic of China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060 Hubei Province People's Republic of China
| |
Collapse
|
37
|
Abnormal Expression of BTLA and CTLA-4 Immune Checkpoint Molecules in Chronic Lymphocytic Leukemia Patients. J Immunol Res 2020; 2020:6545921. [PMID: 32775467 PMCID: PMC7407019 DOI: 10.1155/2020/6545921] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/07/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the peripheral accumulation of neoplastic B cells and is frequently complicated by the systemic immunosuppression associated with an impairment in B and T lymphocyte activation. We hypothesized that the expression of immune checkpoint suppressors B and T lymphocyte attenuator (BTLA) and cytotoxic T lymphocyte antigen (CTLA-4) is disturbed in both lymphocyte subpopulations in CLL. The expression of CTLA-4 and BTLA mRNA was determined by real-time PCR, while CTLA-4 protein expression (surface or intracellular) was estimated in BTLA+ lymphocytes by flow cytometry. In CLL patients, we observed a higher gene transcript level of BTLA and CTLA-4 than in healthy individuals in both freshly isolated and PMA stimulated B and T cells. Remarkably, lower amounts of both inhibitory proteins were found in peripheral blood (PB) CLL B cells, whereas normal BTLA and elevated CTLA-4 were found in T cells. Consistently, there was a prevalence of CTLA-4+ cells within circulating BTLA+ T cells cells of patients confronting PB healthy cells. After in vitro stimulation, the only change found in CLL patients was a decrease in BTLA expression in B and T lymphocytes. In contrast, healthy lymphocytes responded more vigorously as regards the BTLA and CTLA expression with substantially higher frequency of CD69+ cells under the stimulating condition compared to corresponding cells from the CLL group. Our results indicate that CLL development is associated with the affected expression of BTLA and CTLA-4 checkpoint receptors in PB and its impaired expression might be associated with lowering of the threshold for B cell activation and proliferation, while upregulated CTLA-4 expression in CLL peripheral BTLA+ T cells may contribute to suppressed T cell effector functions. This hypothesis needs to be validated in future studies, which would allow us to explain how the increased or decreased expression of these molecules affects the cell function.
Collapse
|
38
|
Chen H, Moussa M, Catalfamo M. The Role of Immunomodulatory Receptors in the Pathogenesis of HIV Infection: A Therapeutic Opportunity for HIV Cure? Front Immunol 2020; 11:1223. [PMID: 32714317 PMCID: PMC7343933 DOI: 10.3389/fimmu.2020.01223] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune activation is the hallmark of HIV infection and plays a role in the pathogenesis of the disease. In the context of suppressed HIV RNA replication by combination antiretroviral therapy (cART), there remains immune activation which is associated to the HIV reservoirs. Persistent virus contributes to a sustained inflammatory environment promoting accumulation of "activated/exhausted" T cells with diminished effector function. These T cells show increased expression of immunomodulatory receptors including Programmed cell death protein (PD1), Cytotoxic T Lymphocyte Associated Protein 4 (CTLA4), Lymphocyte activation gene 3 (LAG3), T cell immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin and mucin domain containing 3 (TIM3) among others. More importantly, recent reports had demonstrated that, HIV infected T cells express checkpoint receptors, contributing to their survival and promoting maintenance of the viral reservoir. Therapeutic strategies are focused on viral reservoir elimination and/or those to achieve sustained cART-free virologic remission. In this review, we will discuss the immunological basis and the latest advances of the use of checkpoint inhibitors to treat HIV infection.
Collapse
Affiliation(s)
- Hui Chen
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
- CMRS/Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Maha Moussa
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| | - Marta Catalfamo
- Department of Microbiology and Immunology, Georgetown University School of Medicine, Washington, DC, United States
| |
Collapse
|
39
|
Pilipow K, Darwich A, Losurdo A. T-cell-based breast cancer immunotherapy. Semin Cancer Biol 2020; 72:90-101. [PMID: 32492452 DOI: 10.1016/j.semcancer.2020.05.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/22/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy has witnessed a new renaissance with the advent of immune checkpoint inhibitors, which reactivate T cells and foster endogenous anti-tumor responses. The excellent results of immunotherapy in the field of melanoma, renal cancer, lung cancer, and other cancer types that have traditionally been known to be immunogenic, rekindled the interest of the oncology community in extending the benefits to all cancers including breast cancer (BC). In this review, we highlight the current state of using T cells as both markers for clinical practice and therapeutic options for BC.
Collapse
Affiliation(s)
- Karolina Pilipow
- Laboratory of Translational Immunology, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy
| | - Abbass Darwich
- Laboratory of Mucosal Immunology and Microbiota, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy; Humanitas University, Department of Biomedical Sciences, Pieve Emanuele, MI, Italy
| | - Agnese Losurdo
- Laboratory of Translational Immunology, Italy; Medical Oncology and Hematology Unit, Italy; Humanitas Clinical and Research Center - IRCCS - Rozzano, MI, Italy.
| |
Collapse
|
40
|
Zhang J, Deng Y, Khoo BL. Fasting to enhance Cancer treatment in models: the next steps. J Biomed Sci 2020; 27:58. [PMID: 32370764 PMCID: PMC7201989 DOI: 10.1186/s12929-020-00651-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
Short-term fasting (STF) is a technique to reduce nutrient intake for a specific period. Since metabolism plays a pivotal role in tumor progression, it can be hypothesized that STF can improve the efficacy of chemotherapy. Recent studies have demonstrated the efficacy of STF in cell and animal tumor models. However, large-scale clinical trials must be conducted to verify the safety and effectiveness of these diets. In this review, we re-examine the concept of how metabolism affects pathophysiological pathways. Next, we provided a comprehensive discussion of the specific mechanisms of STF on tumor progression, derived through studies carried out with tumor models. There are currently at least four active clinical trials on fasting and cancer treatment. Based on these studies, we highlight the potential caveats of fasting in clinical applications, including the onset of metabolic syndrome and other metabolic complications during chemotherapy, with a particular focus on the regulation of the epithelial to mesenchymal pathway and cancer heterogeneity. We further discuss the advantages and disadvantages of the current state-of-art tumor models for assessing the impact of STF on cancer treatment. Finally, we explored upcoming fasting strategies that could complement existing chemotherapy and immunotherapy strategies to enable personalized medicine. Overall, these studies have the potential for breakthroughs in cancer management.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong.
| |
Collapse
|
41
|
Boegeholz J, Brueggen CS, Pauli C, Dimitriou F, Haralambieva E, Dummer R, Manz MG, Widmer CC. Challenges in diagnosis and management of neutropenia upon exposure to immune-checkpoint inhibitors: meta-analysis of a rare immune-related adverse side effect. BMC Cancer 2020; 20:300. [PMID: 32290812 PMCID: PMC7155336 DOI: 10.1186/s12885-020-06763-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 03/17/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cancer immunotherapy via immune-checkpoint inhibition (ICI) by antibodies against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and cell death protein 1 (PD-1) have significantly improved the outcome of metastasized melanoma and of a rapidly increasing number of other cancer types. The anti-tumor effect is often accompanied by immune-related adverse events (irAE). Hematological irAE, specifically neutropenia, are rarely observed. However, neutropenia is associated with high morbidity and mortality due to infection complications. Thus, early detection and treatment is crucial. METHODS We present the clinical course of two patients with severe neutropenia after ICI therapy and demonstrate the difficulty of the diagnosis when a comedication of metamizole, a well-known analgesic drug used to treat cancer pain, is present. Further, we provide a comprehensive descriptive and statistical analysis of published data on diagnostics, treatment and infection complication in patients with at least grade 4 neutropenia by a systematic database search. RESULTS Finally, 34 patients were analyzed, including the two case reports from our cohort. The median onset of neutropenia was 10.5 weeks after first ICI administration (interquartile range: 6 weeks). In 76% (N = 26), a normalization of the neutrophil count was achieved after a median duration of neutropenia of 13 days. In a subsample of 22 patients with detailed data, the infection rate was 13%, proven by positive blood culture in 3 cases, but 68% (N = 15) presented with fever > 38 °C. Treatment regime differed relevantly, but mainly included G-CSF and intravenous corticosteroids. Death was reported in 14 patients (41%), 3 of whom (9%) were associated with hematological irAE but only two directly associated with neutropenia. CONCLUSION With an increasing number of cancer patients eligible to ICI therapy, the incidence of severe hematological toxicities may rise substantially over the next years. Clinicians working in the field of cancer immune therapies should be aware of neutropenia as irAE to provide immediate treatment.
Collapse
Affiliation(s)
- J Boegeholz
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - C S Brueggen
- Department of Dermatology, University and University Hospital Zurich, Zurich, Switzerland
| | - C Pauli
- Department of Dermatology, University and University Hospital Zurich, Zurich, Switzerland
| | - F Dimitriou
- Department of Dermatology, University and University Hospital Zurich, Zurich, Switzerland
| | - E Haralambieva
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, Zurich, Switzerland
| | - R Dummer
- Department of Dermatology, University and University Hospital Zurich, Zurich, Switzerland
| | - M G Manz
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - C C Widmer
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
42
|
Shin EC. Coalition Forces of Immunologists and Oncologists for Defeating Cancer. Immune Netw 2020; 20:e1. [PMID: 32158589 PMCID: PMC7049580 DOI: 10.4110/in.2020.20.e1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 02/18/2020] [Indexed: 02/08/2023] Open
Affiliation(s)
- Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
43
|
Gandhi S, Pandey M, Ammannagari N, Wang C, Bucsek MJ, Hamad L, Repasky E, Ernstoff MS. Impact of concomitant medication use and immune-related adverse events on response to immune checkpoint inhibitors. Immunotherapy 2020; 12:141-149. [PMID: 32064978 DOI: 10.2217/imt-2019-0064] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aim: Patients receiving checkpoint inhibitors (CPI) are frequently on other medications for co-morbidities. We explored the impact of concomitant medication use on outcomes. Materials & methods: 210 metastatic cancer patients on CPI were identified and association between concomitant medication use and immune-related adverse events with clinical outcomes was determined. Results: Aspirin, metformin, β-blockers and statins were not shown to have any statistically significant difference on clinical benefit. 26.3% patients with clinical benefit developed rash versus 11.8% without clinical benefit (p < 0.05) on multivariate analysis. Conclusion: Use of common prescription and nonprescription medications in patients with multiple co-morbidities appears safe and does not have an adverse effect on CPI efficacy. The presence of rash predicted for a better response.
Collapse
Affiliation(s)
- Shipra Gandhi
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Manu Pandey
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Nischala Ammannagari
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Chong Wang
- Department of Biostatistics, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Mark J Bucsek
- Department of Immunology & Cell Stress & Biophysical Therapies Program, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Lamya Hamad
- Melanoma Program, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Clinical Pharmacy Service, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Elizabeth Repasky
- Department of Immunology & Cell Stress & Biophysical Therapies Program, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Marc S Ernstoff
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA.,Melanoma Program, Roswell Park Comprehensive Cancer Center, Elm & Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
44
|
Guerrouahen BS, Maccalli C, Cugno C, Rutella S, Akporiaye ET. Reverting Immune Suppression to Enhance Cancer Immunotherapy. Front Oncol 2020; 9:1554. [PMID: 32039024 PMCID: PMC6985581 DOI: 10.3389/fonc.2019.01554] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/23/2019] [Indexed: 01/26/2023] Open
Abstract
Tumors employ strategies to escape immune control. The principle aim of most cancer immunotherapies is to restore effective immune surveillance. Among the different processes regulating immune escape, tumor microenvironment-associated soluble factors, and/or cell surface-bound molecules are mostly responsible for dysfunctional activity of tumor-specific CD8+T cells. These dynamic immunosuppressive networks prevent tumor rejection at several levels, limiting also the success of immunotherapies. Nevertheless, the recent clinical development of immune checkpoint inhibitors or of molecules modulating cellular targets and immunosuppressive enzymes highlights the great potential of approaches based on the selective disruption of immunosuppressive networks. Currently, the administration of different categories of immunotherapy in combination regimens is the ultimate modality for impacting the survival of cancer patients. With the advent of immune checkpoint inhibitors, designed to mount an effective antitumor immune response, profound changes occurred in cancer immunotherapy: from a global stimulation of the immune system to a specific targeting of an immune component. This review will specifically highlight the players, the mechanisms limiting an efficient antitumor response and the current immunotherapy modalities tailored to target immune suppressive pathways. We also discuss the ongoing challenges encountered by these strategies and provide suggestions for circumventing hurdles to new immunotherapeutic approaches, including the use of relevant biomarkers in the optimization of immunotherapy regimens and the identification of patients who can benefit from defined immune-based approaches.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Cristina Maccalli
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Chiara Cugno
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Emmanuel T Akporiaye
- Veana Therapeutics, Inc., Portland, OR, United States.,Providence Cancer Center, Portland, OR, United States
| |
Collapse
|
45
|
Yoshida K, Okamoto M, Sasaki J, Kuroda C, Ishida H, Ueda K, Ideta H, Kamanaka T, Sobajima A, Takizawa T, Tanaka M, Aoki K, Uemura T, Kato H, Haniu H, Saito N. Anti-PD-1 antibody decreases tumour-infiltrating regulatory T cells. BMC Cancer 2020; 20:25. [PMID: 31914969 PMCID: PMC6950856 DOI: 10.1186/s12885-019-6499-y] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/24/2019] [Indexed: 12/31/2022] Open
Abstract
Background There are many types of therapies for cancer. In these days, immunotherapies, especially immune checkpoint inhibitors, are focused on. Though many types of immune checkpoint inhibitors are there, the difference of effect and its mechanism are unclear. Some reports suggest the response rate of anti-PD-1 antibody is superior to that of anti-PD-L1 antibody and could potentially produce different mechanisms of action. On the other hand, Treg also express PD-1; however, their relationship remains unclear. Methods In this study, we used osteosarcoma cell lines in vitro and osteosarcoma mouse model in vivo. In vitro, we analyzed the effect of IFNγ for expression of PD-L1 on the surface of cell lines by flowcytometry. In vivo, murine osteosarcoma cell line LM8 was subcutaneously transplanted into the dorsum of mice. Mouse anti-PD-1 antibody was intraperitoneally administered. we analysed the effect for survival of anti-PD-1 antibody and proportion of T cells in the tumour by flowcytometry. Results We discovered that IFNγ increased PD-L1 expression on the surface of osteosarcoma cell lines. In assessing the relationship between anti-PD-1 antibody and Treg, we discovered the administration of anti-PD-1 antibody suppresses increases in tumour volume and prolongs overall survival time. In the tumour microenvironment, we found that the administration of anti-PD-1 antibody decreased Treg within the tumour and increased tumour-infiltrating lymphocytes. Conclusions Here we clarify for the first time an additional mechanism of anti-tumour effect—as exerted by anti-PD-1 antibody decreasing Treg— we anticipate that our findings will lead to the development of new methods for cancer treatment.
Collapse
Affiliation(s)
- Kazushige Yoshida
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Masanori Okamoto
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan.
| | - Jun Sasaki
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Chika Kuroda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan
| | - Haruka Ishida
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan
| | - Hirokazu Ideta
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takayuki Kamanaka
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Atsushi Sobajima
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takashi Takizawa
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Manabu Tanaka
- Department of Orthopedic Surgery, Okaya City Hospital, Okaya, Japan
| | - Kaoru Aoki
- Physical Therapy Division, School of Health Sciences, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takeshi Uemura
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan
| | - Hiroyuki Kato
- Department of Orthopaedic Surgery, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Asahi 3-1-1, Matsumoto, 390-8621, Japan.
| |
Collapse
|
46
|
Triaca V, Carito V, Fico E, Rosso P, Fiore M, Ralli M, Lambiase A, Greco A, Tirassa P. Cancer stem cells-driven tumor growth and immune escape: the Janus face of neurotrophins. Aging (Albany NY) 2019; 11:11770-11792. [PMID: 31812953 PMCID: PMC6932930 DOI: 10.18632/aging.102499] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 11/17/2019] [Indexed: 05/12/2023]
Abstract
Cancer Stem Cells (CSCs) are self-renewing cancer cells responsible for expansion of the malignant mass in a dynamic process shaping the tumor microenvironment. CSCs may hijack the host immune surveillance resulting in typically aggressive tumors with poor prognosis.In this review, we focus on neurotrophic control of cellular substrates and molecular mechanisms involved in CSC-driven tumor growth as well as in host immune surveillance. Neurotrophins have been demonstrated to be key tumor promoting signaling platforms. Particularly, Nerve Growth Factor (NGF) and its specific receptor Tropomyosin related kinase A (TrkA) have been implicated in initiation and progression of many aggressive cancers. On the other hand, an active NGF pathway has been recently proven to be critical to oncogenic inflammation control and in promoting immune response against cancer, pinpointing possible pro-tumoral effects of NGF/TrkA-inhibitory therapy.A better understanding of the molecular mechanisms involved in the control of tumor growth/immunoediting is essential to identify new predictive and prognostic intervention and to design more effective therapies. Fine and timely modulation of CSCs-driven tumor growth and of peripheral lymph nodes activation by the immune system will possibly open the way to precision medicine in neurotrophic therapy and improve patient's prognosis in both TrkA- dependent and independent cancers.
Collapse
Affiliation(s)
- Viviana Triaca
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), International Campus A. Buzzati-Traverso, Monterotondo Scalo, Rome, Italy
| | - Valentina Carito
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Elena Fico
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Pamela Rosso
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Massimo Ralli
- Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | | | - Antonio Greco
- Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| | - Paola Tirassa
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), at Department of Sense Organs, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
47
|
Martins PR, Machado CMT, Coxir SA, de Oliveira AJ, Moreira TB, Campos LS, Alcântara R, de Paula SOC, de Oliveira Salles PG, Gollob KJ, Magalhães WCS. Cervical cancer patients that respond to chemoradiation therapy display an intense tumor infiltrating immune profile before treatment. Exp Mol Pathol 2019; 111:104314. [DOI: 10.1016/j.yexmp.2019.104314] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023]
|
48
|
Targeting Immune-Related Biological Processes in Solid Tumors: We do Need Biomarkers. Int J Mol Sci 2019; 20:ijms20215452. [PMID: 31683784 PMCID: PMC6862285 DOI: 10.3390/ijms20215452] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has become the standard-of-care in many solid tumors. Despite the significant recent achievements in the diagnosis and treatment of cancer, several issues related to patients’ selection for immunotherapy remain unsolved. Multiple lines of evidence suggest that, in this setting, the vision of a single biomarker is somewhat naïve and imprecise, given that immunotherapy does not follow the rules that we have experienced in the past for targeted therapies. On the other hand, additional immune-related biomarkers that are reliable in real-life clinical practice remain to be identified. Recently, the immune-checkpoint blockade has been approved in the US irrespective of the tumor site of origin. Further histology-agnostic approvals, coupled with with tumor-specific companion diagnostics and guidelines, are expected in this field. In addition, immune-related biomarkers can also have a significant prognostic value. In this review, we provide an overview of the role of these biomarkers and their characterization in the management of lung cancer, melanoma, colorectal cancer, gastric cancer, head and neck cancer, renal cell carcinoma, urothelial cancers, and breast cancer.
Collapse
|
49
|
Chakraborty D, Pati S, Bose S, Dhar S, Dutta S, Sa G. Cancer immunotherapy: present scenarios and the future of immunotherapy. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
50
|
Bang YJ, Kang YK, Catenacci DV, Muro K, Fuchs CS, Geva R, Hara H, Golan T, Garrido M, Jalal SI, Borg C, Doi T, Yoon HH, Savage MJ, Wang J, Dalal RP, Shah S, Wainberg ZA, Chung HC. Pembrolizumab alone or in combination with chemotherapy as first-line therapy for patients with advanced gastric or gastroesophageal junction adenocarcinoma: results from the phase II nonrandomized KEYNOTE-059 study. Gastric Cancer 2019; 22:828-837. [PMID: 30911859 PMCID: PMC6570680 DOI: 10.1007/s10120-018-00909-5] [Citation(s) in RCA: 195] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/01/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The multicohort, phase II, nonrandomized KEYNOTE-059 study evaluated pembrolizumab ± chemotherapy in advanced gastric/gastroesophageal junction cancer. Results from cohorts 2 and 3, evaluating first-line therapy, are presented. METHODS Patients ≥ 18 years old had previously untreated recurrent or metastatic gastric/gastroesophageal junction adenocarcinoma. Cohort 3 (monotherapy) had programmed death receptor 1 combined positive score ≥ 1. Cohort 2 (combination therapy) received pembrolizumab 200 mg on day 1, cisplatin 80 mg/m2 on day 1 (up to 6 cycles), and 5-fluorouracil 800 mg/m2 on days 1-5 of each 3-week cycle (or capecitabine 1000 mg/m2 twice daily in Japan). Primary end points were safety (combination therapy) and objective response rate per Response Evaluation Criteria in Solid Tumors version 1.1 by central review, and safety (monotherapy). RESULTS In the combination therapy and monotherapy cohorts, 25 and 31 patients were enrolled; median follow-up was 13.8 months (range 1.8-24.1) and 17.5 months (range 1.7-20.7), respectively. In the combination therapy cohort, grade 3/4 treatment-related adverse events occurred in 19 patients (76.0%); none were fatal. In the monotherapy cohort, grade 3-5 treatment-related adverse events occurred in seven patients (22.6%); one death was attributed to a treatment-related adverse event (pneumonitis). The objective response rate was 60.0% [95% confidence interval (CI), 38.7-78.9] (combination therapy) and 25.8% (95% CI 11.9-44.6) (monotherapy). CONCLUSIONS Pembrolizumab demonstrated antitumor activity and was well tolerated as monotherapy and in combination with chemotherapy in patients with previously untreated advanced gastric/gastroesophageal junction adenocarcinoma. CLINICAL TRIAL ClinicalTrials.gov NCT02335411.
Collapse
Affiliation(s)
- Yung-Jue Bang
- Department of Internal Medicine, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Yoon-Koo Kang
- Division of Oncology, Department of Internal Medicine, Asan Medical Center, University of Ulsan, Seoul, Republic of Korea
| | - Daniel V Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL, USA
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Charles S Fuchs
- Department of Medical Oncology, Yale Cancer Center, New Haven, CT, USA
| | - Ravit Geva
- Department of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Hiroki Hara
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Talia Golan
- Department of Oncology, The Oncology Institute at the Chaim Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo Garrido
- Hematology and Medical Oncology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shadia I Jalal
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christophe Borg
- Department of Medical Oncology, University Hospital of Besançon, Besancon, France
| | - Toshihiko Doi
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Kashiwa, Japan
| | - Harry H Yoon
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Mary J Savage
- Medical Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Jiangdian Wang
- Medical Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Rita P Dalal
- Medical Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Sukrut Shah
- Medical Oncology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Zev A Wainberg
- Division of Hematology Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Hyun Cheol Chung
- Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|