1
|
Robison B, Diong SJ, Kumar A, Moon TM, Chang O, Chau B, Bee C, Barman I, Rajpal A, Korman AJ, West S, Strop P, Lee PS. Engineered ipilimumab variants that bind human and mouse CTLA-4. MAbs 2025; 17:2451296. [PMID: 39849917 PMCID: PMC11776466 DOI: 10.1080/19420862.2025.2451296] [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: 10/24/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/25/2025] Open
Abstract
Testing of candidate monoclonal antibody therapeutics in preclinical models is an essential step in drug development. Identification of antibody therapeutic candidates that bind their human targets and cross-react to mouse orthologs is often challenging, especially for targets with low sequence homology. In such cases, surrogate antibodies that bind mouse orthologs must be used. The antibody 9D9, which binds mouse CTLA-4, is a commonly used surrogate for CTLA-4 checkpoint blockade studies in mouse cancer models. In this work, we reveal that 9D9 has significant biophysical dissimilarities to therapeutic CTLA-4 antibodies. The 9D9-mCTLA4 complex crystal structure was determined and shows that the surrogate antibody binds an epitope distinct from ipilimumab and tremelimumab. In addition, while ipilimumab has pH-independent binding to hCTLA-4, 9D9 loses binding to mCTLA-4 at physiologically relevant acidic pH ranges. We used phage and yeast display to engineer ipilimumab to bind mouse CTLA-4 with single-digit nM affinity from an initial state with no apparent binding. The engineered variants showed pH-independent and cross-reactive binding to both mouse and human CTLA-4. Crystal structures of a variant in complex with both mouse and human CTLA-4 confirmed that it targets an equivalent epitope as ipilimumab. These cross-reactive ipilimumab variants may facilitate improved translatability and future mechanism-of-action studies for anti-CTLA-4 targeting in murine models.
Collapse
Affiliation(s)
- Brett Robison
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - SJ Diong
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Anusha Kumar
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Thomas M. Moon
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Olin Chang
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Bryant Chau
- Large Molecule Drug Discovery, Genentech, Research and Early Development, South San Francisco, CA, USA
| | - Christine Bee
- Biochemistry and Biophysics, Merck, South San Francisco, CA, USA
| | - Ishita Barman
- Protein and Antibody Portfolio, Genscript, South San Francisco, CA, USA
| | | | | | - Sean West
- Discovery Biotherapeutics, Bristol Myers Squibb, Redwood City, CA, USA
| | - Pavel Strop
- Research, Tallac Therapeutics, Burlingame, CA, USA
| | - Peter S. Lee
- Biotherapeutics and Genetic Medicine, AbbVie, South San Francisco, CA, USA
| |
Collapse
|
2
|
Xu P, Sun X, Pan L, Zhu J, Qian S. Disulfidptosis-related LncRNAs forecast the prognosis of acute myeloid leukemia. Sci Rep 2025; 15:13635. [PMID: 40254646 PMCID: PMC12009979 DOI: 10.1038/s41598-025-95607-5] [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: 07/24/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematologic malignancy with a poor prognosis for patients. Disulfidptosis response-related long non-coding RNAs (DRLs) have been demonstrated to be closely associated with cancer development. Therefore, this study aims to construct a prognostic DRL signature and investigate the immune microenvironment for AML. RNA-seq and clinical data for AML patients were obtained from The Cancer Genome Atlas (TCGA) database. A total of 344 disulfidptosis-associated lncRNAs were identified, and a prognostic model consisting of seven lncRNAs was constructed and validated. Two risk groups, high-risk and low-risk, were identified. The model demonstrated a robust capacity to predict prognosis, with a worse overall survival for patients in the high-risk group. Additionally, differential expression of the seven lncRNAs were relatively higher in AML samples than in control samples via quantitative polymerase chain reaction(qPCR). The Kyoto Encyclopedia of Genes and Genomes (KEGG) and immune infiltration analysis revealed a substantial infiltration of immune cells and enrichment of immune pathways in the high-risk group. The sensitivity of AML patients to drugs varied according to their risk grade. This study identified a DRL signature, which can effectively predict the prognosis of AML and better understand the mechanism of disulfidptosis in AML. This provides a basis for personalized immunotherapy in AML patients.
Collapse
Affiliation(s)
- Pei Xu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Xiaolin Sun
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Lingxiao Pan
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China
| | - Jianfeng Zhu
- Department of Hematology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, China.
| | - Sixuan Qian
- Department of Hematology, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210003, China.
| |
Collapse
|
3
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [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: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
4
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
5
|
Chand D, Savitsky DA, Krishnan S, Mednick G, Delepine C, Garcia-Broncano P, Soh KT, Wu W, Wilkens MK, Udartseva O, Vincent S, Joshi B, Keith JG, Manrique M, Marques M, Tanne A, Levey DL, Han H, Ng S, Ridpath J, Huber O, Morin B, Galand C, Bourdelais S, Gombos RB, Ward R, Qin Y, Waight JD, Costa MR, Sebastian-Yague A, Rudqvist NP, Pupecka-Swider M, Venkatraman V, Slee A, Patel JM, Grossman JE, Wilson NS, Von Hoff DD, Stebbing J, Curiel TJ, Buell JS, O’Day SJ, Stein RB. Botensilimab, an Fc-Enhanced Anti-CTLA-4 Antibody, Is Effective against Tumors Poorly Responsive to Conventional Immunotherapy. Cancer Discov 2024; 14:2407-2429. [PMID: 39083809 PMCID: PMC11609826 DOI: 10.1158/2159-8290.cd-24-0190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/02/2024]
Abstract
SIGNIFICANCE This study reveals that Fc-enhanced anti-CTLA-4 harnesses novel mechanisms to overcome the limitations of conventional anti-CTLA-4, effectively treating poorly immunogenic and treatment-refractory cancers. Our findings support the development of a new class of immuno-oncology agents, capable of extending clinical benefit to patients with cancers resistant to current immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wei Wu
- Agenus Inc, Lexington, Massachusetts
| | | | | | | | | | | | | | | | | | | | - Haiyong Han
- The Translational Genomics Research Institute, Phoenix, Arizona
| | - Serina Ng
- The Translational Genomics Research Institute, Phoenix, Arizona
| | | | | | | | | | | | | | | | - Yu Qin
- Agenus Inc, Lexington, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | - Tyler J. Curiel
- Agenus Inc, Lexington, Massachusetts
- Dartmouth Cancer Center and the Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - Jennifer S. Buell
- Agenus Inc, Lexington, Massachusetts
- MiNK Therapeutics, Lexington, Massachusetts
| | | | - Robert B. Stein
- Agenus Inc, Lexington, Massachusetts
- MiNK Therapeutics, Lexington, Massachusetts
| |
Collapse
|
6
|
Chen L, Huang L, Gu Y, Li C, Sun P, Xiang Y. Novel post-translational modifications of protein by metabolites with immune responses and immune-related molecules in cancer immunotherapy. Int J Biol Macromol 2024; 277:133883. [PMID: 39033895 DOI: 10.1016/j.ijbiomac.2024.133883] [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: 07/01/2023] [Revised: 06/30/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Tumour immunotherapy is an effective and essential treatment for cancer. However, the heterogeneity of tumours and the complex and changeable tumour immune microenvironment (TME) creates many uncertainties in the clinical application of immunotherapy, such as different responses to tumour immunotherapy and significant differences in individual efficacy. It makes anti-tumour immunotherapy face many challenges. Immunometabolism is a critical determinant of immune cell response to specific immune effector molecules, significantly affecting the effects of tumour immunotherapy. It is attributed mainly to the fact that metabolites can regulate the function of immune cells and immune-related molecules through the protein post-translational modifications (PTMs) pathway. This study systematically summarizes a variety of novel protein PTMs including acetylation, propionylation, butyrylation, succinylation, crotonylation, malonylation, glutarylation, 2-hydroxyisobutyrylation, β-hydroxybutyrylation, benzoylation, lactylation and isonicotinylation in the field of tumour immune regulation and immunotherapy. In particular, we elaborate on how different PTMs in the TME can affect the function of immune cells and lead to immune evasion in cancer. Lastly, we highlight the potential treatment with the combined application of target-inhibited protein modification and immune checkpoint inhibitors (ICIs) for improved immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China
| | - Yu Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Chen Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China.
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China.
| |
Collapse
|
7
|
Shen S, Hong Y, Huang J, Qu X, Sooranna SR, Lu S, Li T, Niu B. Targeting PD-1/PD-L1 in tumor immunotherapy: Mechanisms and interactions with host growth regulatory pathways. Cytokine Growth Factor Rev 2024; 79:16-28. [PMID: 39179486 DOI: 10.1016/j.cytogfr.2024.08.001] [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: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Songyu Shen
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Yihan Hong
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Jiajun Huang
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Xiaosheng Qu
- Guangxi Botanical Garden of Medicinal Plants, Nanning, Guangxi 530023, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, 369 Fulham Road, London SW10 9NH, United Kingdom
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 169 Changle West Rd, Xi'an 710032, China.
| | - Bing Niu
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China.
| |
Collapse
|
8
|
Xu Z, Zhang H, Ma G, Meng W, Du J, Wu X, Yang B, Wang N, Ding Y, Zhang Q, Li N, Zhang X, Yu G, Liu S, Li Z. Real‑world evidence of advanced non‑small cell lung carcinoma treated with an immune checkpoint inhibitor plus chemotherapy. Oncol Lett 2024; 28:405. [PMID: 38983127 PMCID: PMC11228919 DOI: 10.3892/ol.2024.14538] [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: 02/22/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Immunotherapy is an effective treatment strategy for patients with advanced non-small cell lung cancer (NSCLC). Although clinical trials on immunotherapy have provided promising results, real-world research in clinical practice is needed to assess the effectiveness and safety of immunotherapy. The present study aimed to characterize real-world outcomes in patients with advanced NSCLC treated with immune checkpoint inhibitor (ICI)-based regimens. The medical records of patients with advanced NSCLC, who were treated with programmed cell death protein-1 (PD-1)/programmed cell death 1 ligand 1 (PD-L1) inhibitors, were reviewed for data collection. The primary objectives were to evaluate progression-free survival (PFS) and overall survival (OS). Therefore, multiple Cox regression models were used to investigate the predictive factors for survival outcomes. Furthermore, survival curves for PFS and OS were created using Kaplan-Meier estimates and compared using the log-rank test. The present study included a total of 133 patients with advanced NSCLC who received therapy with ICIs between January 1, 2019 and December 31, 2022. The final follow-up date was August 24, 2023. The median PFS and OS times were 9.8 and 27.2 months, respectively. Univariate Cox regression analysis demonstrated that sex, clinical stage, PD-L1 status, previous systemic therapy, and brain and liver metastases were associated with PFS, while Eastern Cooperative Oncology Group (ECOG) status, clinical stage, PD-L1 status and brain metastasis were associated with OS. Furthermore, multivariate Cox regression analysis demonstrated that a PD-L1 tumor proportion score (TPS) of ≥50% was an indicator of favorable PFS and OS. An ECOG performance status score of ≥1 was also associated with poor OS but not with PFS. Furthermore, brain metastasis was an indicator for poor PFS and OS, while liver metastasis was only associated with a poor PFS. Finally, the results of the present study demonstrated that PD-L1 status was an independent predictor for PFS and OS in patients with advanced NSCLC, especially adenocarcinoma, who were treated with ICIs plus chemotherapy. The results also suggested that patients with a PD-L1 TPS of ≥50% could benefit when the aforementioned regimens were administrated as a first-line or later-line therapy.
Collapse
Affiliation(s)
- Zihan Xu
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
- Department of Pneumology, Sunshine Union Hospital, Weifang, Shandong 261000, P.R. China
| | - Huien Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Guikai Ma
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Wenjuan Meng
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Junliang Du
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Xin Wu
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Baohong Yang
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Ningning Wang
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Yanhong Ding
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Qingyun Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Na Li
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Xuede Zhang
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Guohua Yu
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Shuzhen Liu
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
| | - Zhenhua Li
- Department of Oncology, The First Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong 261000, P.R. China
- Department of Pathology, Shanghai Clinical Research and Trial Center, Shanghai 201203, P.R. China
| |
Collapse
|
9
|
Rizzotto E, Inciardi I, Fongaro B, Trolese P, Miolo G, Polverino de Laureto P. Light exacerbates local and global effects induced by pH unfolding of Ipilimumab. Eur J Pharm Biopharm 2024; 201:114387. [PMID: 38944210 DOI: 10.1016/j.ejpb.2024.114387] [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: 04/05/2024] [Revised: 05/30/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Monoclonal antibodies (mAbs) are an essential class of therapeutic proteins for the treatment of cancer, autoimmune and rare diseases. During their production, storage, and administration processes, these proteins encounter various stressors such as temperature fluctuations, vibrations, and light exposure, able to induce chemico-physical modifications to their structure. Viral inactivation is a key step in downstream processes, and it is achieved by titration of the mAb at low pH, followed by neutralization. The changes of the pH pose a significant risk of unfolding and subsequent aggregation to proteins, thereby affecting their manufacturing. This study aims to investigate whether a combined exposure to light during the viral inactivation process can further affect the structural integrity of Ipilimumab, a mAb primarily used in the treatment of metastatic melanoma. The biophysical and biochemical characterization of Ipilimumab revealed that pH variation is a considerable risk for its stability with irreversible unfolding at pH 2. The threshold for Ipilimumab denaturation lies between pH 2 and 3 and is correlated with the loss of the protein structural cooperativity, which is the most critical factor determining the protein refolding. Light has demonstrated to exacerbate some local and global effects making pH-induced exposed regions more vulnerable to structural and chemical changes. Therefore, specific precautions to real-life exposure to ambient light during the sterilization process of mAbs should be considered to avoid loss of the therapeutic activity and to increase the yield of production. Our findings underscore the critical role of pH optimization in preserving the structural integrity and therapeutic efficacy of mAbs. Moreover, a detailed conformational study on the structural modifications of Ipilimumab may improve the chemico-physical knowledge of this effective drug and suggest new production strategies for more stable products under some kind of stress conditions.
Collapse
Affiliation(s)
- Elena Rizzotto
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 30131 Padova, Italy
| | - Ilenia Inciardi
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 30131 Padova, Italy
| | - Benedetta Fongaro
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 30131 Padova, Italy
| | - Philipp Trolese
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 30131 Padova, Italy
| | - Giorgia Miolo
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo 5, 30131 Padova, Italy
| | | |
Collapse
|
10
|
Younis A, Gribben J. Immune Checkpoint Inhibitors: Fundamental Mechanisms, Current Status and Future Directions. IMMUNO 2024; 4:186-210. [DOI: 10.3390/immuno4030013] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) are a promising form of immunotherapy that have significantly changed the therapeutic landscape for many advanced cancers. They have shown unique clinical benefit against a broad range of tumour types and a strong overall impact on survival in studied patient populations. However, there are still many limitations holding back this immunotherapy from reaching its full potential as a possible curative option for advanced cancer patients. A great deal of research is being undertaken in the hope of driving advancements in this area, building a better understanding of the mechanisms behind immune checkpoint inhibition and ultimately developing more effective, safer, and wider-reaching agents. Taking into account the current literature on this topic, this review aims to explore in depth the basis of the use of ICIs in the treatment of advanced cancers, evaluate its efficacy and safety, consider its current limitations, and finally reflect on what the future holds for this very promising form of cancer immunotherapy.
Collapse
Affiliation(s)
- Abdullah Younis
- Barts and the London School of Medicine and Dentistry, London E1 2AD, UK
| | - John Gribben
- Barts Cancer Institute, Queen Mary University of London, London EC1M 6AU, UK
| |
Collapse
|
11
|
Afshari AR, Sanati M, Ahmadi SS, Kesharwani P, Sahebkar A. Harnessing the capacity of phytochemicals to enhance immune checkpoint inhibitor therapy of cancers: A focus on brain malignancies. Cancer Lett 2024; 593:216955. [PMID: 38750720 DOI: 10.1016/j.canlet.2024.216955] [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: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/23/2024]
Abstract
Brain cancers, particularly glioblastoma multiforme (GBM), are challenging health issues with frequent unmet aspects. Today, discovering safe and effective therapeutic modalities for brain tumors is among the top research interests. Immunotherapy is an emerging area of investigation in cancer treatment. Since immune checkpoints play fundamental roles in repressing anti-cancer immunity, diverse immune checkpoint inhibitors (ICIs) have been developed, and some monoclonal antibodies have been approved clinically for particular cancers; nevertheless, there are significant concerns regarding their efficacy and safety in brain tumors. Among the various tools to modify the immune checkpoints, phytochemicals show good effectiveness and excellent safety, making them suitable candidates for developing better ICIs. Phytochemicals regulate multiple immunological checkpoint-related signaling pathways in cancer biology; however, their efficacy for clinical cancer immunotherapy remains to be established. Here, we discussed the involvement of immune checkpoints in cancer pathology and summarized recent advancements in applying phytochemicals in modulating immune checkpoints in brain tumors to highlight the state-of-the-art and give constructive prospects for future research.
Collapse
Affiliation(s)
- Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
12
|
Rahmat JN, Liu J, Chen T, Li Z, Zhang Y. Engineered biological nanoparticles as nanotherapeutics for tumor immunomodulation. Chem Soc Rev 2024; 53:5862-5903. [PMID: 38716589 DOI: 10.1039/d3cs00602f] [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: 06/05/2024]
Abstract
Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.
Collapse
Affiliation(s)
- Juwita N Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117585, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, The City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
13
|
AlRubaish FA, Gupta N, Shi MZ, Christopoulos S. Immune checkpoint inhibitor-induced hypophysitis with transient ACTH-dependent hypercortisolism. BMJ Case Rep 2024; 17:e258701. [PMID: 38772867 DOI: 10.1136/bcr-2023-258701] [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] [Indexed: 05/23/2024] Open
Abstract
A woman in her 70s with metastatic melanoma presenting with refractory hypokalaemia on combined immune checkpoint inhibitors, nivolumab-ipilimumab, was diagnosed with adrenocorticotropic hormone (ACTH)-dependent hypercortisolism 11 weeks following the initiation of her immunotherapy. Investigations also demonstrated central hypothyroidism and hypogonadotropic hypogonadism. She underwent imaging studies of her abdomen and brain which revealed normal adrenal glands and pituitary, respectively. She was started on levothyroxine replacement and had close pituitary function monitoring. Two weeks later, her cortisol and ACTH levels started to trend down. She finally developed secondary adrenal insufficiency and was started on hydrocortisone replacement 4 weeks thereafter.This report highlights a case of immunotherapy-related hypophysitis with well-documented transient central hypercortisolism followed, within weeks, by profound secondary adrenal insufficiency. Healthcare professionals should remain vigilant in monitoring laboratory progression in these patients. Early recognition of the phase of hypercortisolism and its likely rapid transformation into secondary adrenal insufficiency can facilitate timely hormonal replacement and prevent complications.
Collapse
Affiliation(s)
- Fatima Abdullah AlRubaish
- Department of Internal Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Eastern Province, Saudi Arabia
| | - Nisha Gupta
- McGill University Health Centre, Montreal, Quebec, Canada
| | - Meng Zhu Shi
- Department of Internal Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | | |
Collapse
|
14
|
Bauer M, Schöbel CM, Wickenhauser C, Seliger B, Jasinski-Bergner S. Deciphering the role of alternative splicing in neoplastic diseases for immune-oncological therapies. Front Immunol 2024; 15:1386993. [PMID: 38736877 PMCID: PMC11082354 DOI: 10.3389/fimmu.2024.1386993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
Alternative splicing (AS) is an important molecular biological mechanism regulated by complex mechanisms involving a plethora of cis and trans-acting elements. Furthermore, AS is tissue specific and altered in various pathologies, including infectious, inflammatory, and neoplastic diseases. Recently developed immuno-oncological therapies include monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells targeting, among others, immune checkpoint (ICP) molecules. Despite therapeutic successes have been demonstrated, only a limited number of patients showed long-term benefit from these therapies with tumor entity-related differential response rates were observed. Interestingly, splice variants of common immunotherapeutic targets generated by AS are able to completely escape and/or reduce the efficacy of mAb- and/or CAR-based tumor immunotherapies. Therefore, the analyses of splicing patterns of targeted molecules in tumor specimens prior to therapy might help correct stratification, thereby increasing therapy success by antibody panel selection and antibody dosages. In addition, the expression of certain splicing factors has been linked with the patients' outcome, thereby highlighting their putative prognostic potential. Outstanding questions are addressed to translate the findings into clinical application. This review article provides an overview of the role of AS in (tumor) diseases, its molecular mechanisms, clinical relevance, and therapy response.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Chiara-Maria Schöbel
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Barbara Seliger
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
- Department of Good Manufacturing Practice (GMP) Development & Advanced Therapy Medicinal Products (ATMP) Design, Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany
- Institute for Medical Immunology, Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Simon Jasinski-Bergner
- Institute for Translational Immunology, Brandenburg Medical School (MHB), Theodor Fontane, Brandenburg an der Havel, Germany
| |
Collapse
|
15
|
Guo Z, Ye J, Cheng X, Wang T, Zhang Y, Yang K, Du S, Li P. Nanodrug Delivery Systems in Antitumor Immunotherapy. Biomater Res 2024; 28:0015. [PMID: 38840653 PMCID: PMC11045275 DOI: 10.34133/bmr.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/26/2024] [Indexed: 06/07/2024] Open
Abstract
Cancer has become one of the most important factors threatening human health, and the global cancer burden has been increasing rapidly. Immunotherapy has become another clinical research hotspot after surgery, chemotherapy, and radiotherapy because of its high efficiency and tumor metastasis prevention. However, problems such as lower immune response rate and immune-related adverse reaction in the clinical application of immunotherapy need to be urgently solved. With the development of nanodrug delivery systems, various nanocarrier materials have been used in the research of antitumor immunotherapy with encouraging therapeutic results. In this review, we mainly summarized the combination of nanodrug delivery systems and immunotherapy from the following 4 aspects: (a) nanodrug delivery systems combined with cytokine therapy to improve cytokines delivery in vivo; (b) nanodrug delivery systems provided a suitable platform for the combination of immune checkpoint blockade therapy with other tumor treatments; (c) nanodrug delivery systems helped deliver antigens and adjuvants for tumor vaccines to enhance immune effects; and (d) nanodrug delivery systems improved tumor treatment efficiency and reduced toxicity for adoptive cell therapy. Nanomaterials chosen by researchers to construct nanodrug delivery systems and their function were also introduced in detail. Finally, we discussed the current challenges and future prospects in combining nanodrug delivery systems with immunotherapy.
Collapse
Affiliation(s)
- Zishuo Guo
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jinhong Ye
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xuehao Cheng
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tieshan Wang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- YiDu Central Hospital of Weifang, Weifang, Shandong 262500, China
| | - Kaili Yang
- Beijing University of Chinese Medicine, Beijing 102488, China
| | | | - Pengyue Li
- Address correspondence to: (P.L.); (S.D.)
| |
Collapse
|
16
|
Wang M, Chen L, He J, Xia W, Ye Z, She J. Structural insights into IL-6 signaling inhibition by therapeutic antibodies. Cell Rep 2024; 43:113819. [PMID: 38393945 DOI: 10.1016/j.celrep.2024.113819] [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: 08/22/2023] [Revised: 12/14/2023] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Antibody inhibitors of the interleukin-6 (IL-6) signaling pathway, such as tocilizumab and sarilumab, have been used to treat rheumatoid arthritis, chimeric antigen receptor T cell-induced cytokine storm, and severe COVID-19 pneumonia. Here, we solve the cryogenic electron microscopy structures of sarilumab and tocilizumab in complex with IL-6R to resolutions of 3.2 and 3.3 Å, respectively. These structures reveal that both tocilizumab and sarilumab bind to the D3 domain of IL-6R. The binding surfaces of the two antibodies largely overlap, but the detailed interactions are different. Functional studies of various mutants show results consistent with our structural analysis of the antibodies and IL-6R interactions. Structural comparisons with the IL-6/IL-6R/gp130 complex indicate that sarilumab and tocilizumab probably inhibit IL-6/IL-6R signaling by competing for the IL-6 binding site. In summary, this work reveals the antibody-blocking mechanism of the IL-6 signaling pathway and paves the way for future antibody discovery.
Collapse
Affiliation(s)
- Mingxing Wang
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Long Chen
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Jin He
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China
| | - Wenqiang Xia
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, Zhejiang, China; College of Agriculture and Biotechnology, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zihong Ye
- Zhejiang Provincial Key Laboratory of Biometrology and Inspection & Quarantine, College of Life Sciences, China Jiliang University, Hangzhou 310018, Zhejiang, China.
| | - Ji She
- MOE Key Laboratory for Cellular Dynamics, School of Life Sciences, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, Anhui, China.
| |
Collapse
|
17
|
Kennedy PT, Saulters EL, Duckworth AD, Lim YJ, Woolley JF, Slupsky JR, Cragg MS, Ward FJ, Dahal LN. Soluble CTLA-4 attenuates T cell activation and modulates anti-tumor immunity. Mol Ther 2024; 32:457-468. [PMID: 38053333 PMCID: PMC10861965 DOI: 10.1016/j.ymthe.2023.11.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/12/2023] [Accepted: 11/30/2023] [Indexed: 12/07/2023] Open
Abstract
CTLA-4 is a crucial immune checkpoint receptor involved in the maintenance of immune homeostasis, tolerance, and tumor control. Antibodies targeting CTLA-4 have been promising treatments for numerous cancers, but the mechanistic basis of their anti-tumoral immune-boosting effects is poorly understood. Although the ctla4 gene also encodes an alternatively spliced soluble variant (sCTLA-4), preclinical/clinical evaluation of anti-CTLA-4-based immunotherapies have not considered the contribution of this isoform. Here, we explore the functional properties of sCTLA-4 and evaluate the efficacy of isoform-specific anti-sCTLA-4 antibody targeting in a murine cancer model. We show that expression of sCTLA-4 by tumor cells suppresses CD8+ T cells in vitro and accelerates growth and experimental metastasis of murine tumors in vivo. These effects were accompanied by modification of the immune infiltrate, notably restraining CD8+ T cells in a non-cytotoxic state. sCTLA-4 blockade with isoform-specific antibody reversed this restraint, enhancing intratumoral CD8+ T cell activation and cytolytic potential, correlating with therapeutic efficacy and tumor control. This previously unappreciated role of sCTLA-4 suggests that the biology and function of multi-gene products of immune checkpoint receptors need to be fully elucidated for improved mechanistic understanding of cancer immunotherapies.
Collapse
Affiliation(s)
- Paul T Kennedy
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, UK
| | - Emma L Saulters
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, UK
| | - Andrew D Duckworth
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L69 3GE Liverpool, UK
| | - Yeong Jer Lim
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L69 3GE Liverpool, UK
| | - John F Woolley
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, UK
| | - Joseph R Slupsky
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, L69 3GE Liverpool, UK
| | - Mark S Cragg
- Centre for Cancer Immunology, University of Southampton, SO16 6YD Southampton, UK
| | - Frank J Ward
- Department of Immunology, University of Aberdeen, AB25 2ZD Aberdeen, UK
| | - Lekh N Dahal
- Department of Pharmacology and Therapeutics, University of Liverpool, L69 3GE Liverpool, UK.
| |
Collapse
|
18
|
Duke S, Maiarana J, Yousefi P, Burks E, Gerrie S, Setiadi A, Hildebrand KJ, James E, Turvey SE, Markle JG, Biggs CM. Expanding the molecular and phenotypic spectrum of CTLA-4 insufficiency. Pediatr Allergy Immunol 2024; 35:e14077. [PMID: 38351878 PMCID: PMC12019878 DOI: 10.1111/pai.14077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Affiliation(s)
- Sean Duke
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
| | - James Maiarana
- Department of Pathology, Microbiology, and Immunology,
Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Pariya Yousefi
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
| | | | - Samantha Gerrie
- Department of Radiology, BC Children’s Hospital, The
University of British Columbia, Vancouver, British Columbia, Canada
| | - Audi Setiadi
- Department of Pathology, BC Children’s Hospital, The
University of British Columbia, Vancouver, British Columbia, Canada
| | - BC Children’s and St Paul’s Hospital Members
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology, BC Children’s Hospital, The
University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, BC Children’s
Hospital, The University of British Columbia, Vancouver, British Columbia,
Canada
- Division of Allergy & Immunology, St Paul’s
Hospital, The University of British Columbia, Vancouver, British Columbia,
Canada
- Division of Rheumatology, Vancouver General Hospital and St
Paul’s Hospital, The University of British Columbia, Vancouver, British
Columbia, Canada
| | - Kyla J. Hildebrand
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
| | - Elliot James
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
| | - Stuart E. Turvey
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
| | - Janet G. Markle
- Department of Pathology, Microbiology, and Immunology,
Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Catherine M. Biggs
- Department of Pediatrics, BC Children’s Hospital,
The University of British Columbia, Vancouver, British Columbia, Canada
- Division of Allergy & Immunology, St Paul’s
Hospital, The University of British Columbia, Vancouver, British Columbia,
Canada
| |
Collapse
|
19
|
Sharma N, Mazumder R, Rai P. Revolutionizing Skin Cancer Treatment: The Rise of PD-1/PDL-1 and CTLA-4 as Key Therapeutic Targets. Curr Drug Targets 2024; 25:1012-1026. [PMID: 39257156 DOI: 10.2174/0113894501320281240822052657] [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: 04/20/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 09/12/2024]
Abstract
Skin cancer is a significant health concern, affecting millions of individuals globally on an annual basis. According to data from the World Health Organization, it stands as the most prevalent form of cancer within the white population. Current treatments for skin cancer typically involve a combination of chemotherapy, radiation therapy, and surgery. However, these methods often come with drawbacks, such as side effects and potential scarring. Therefore, there is a growing need for alternative treatments that can offer effective results with fewer adverse effects, driving ongoing research in skin cancer therapy. The advancement of immune checkpoint inhibitors has been facilitated by a more profound comprehension of the interplay between tumors and the immune system, along with the regulatory mechanisms governing T-cells. As cancer treatment continues to evolve, immunotherapy is emerging as a powerful strategy, leading to a growing interest in the role of immunological checkpoints in skin cancer. Various types of immune checkpoints and their expression, including PD-1, PDL-1, CTLA-4, lymphocyte activation gene 3, and B7-H3, along with their blockers and monoclonal antibodies, have been established for various cancers. PD-1, PDL-1, and CTLA-4 are crucial immune system regulators, acting as brakes to prevent T-- cell overactivation and potential autoimmunity. However, tumors can exploit these checkpoints to evade immune detection. Inhibiting these immune checkpoints can enhance the body's ability to recognize and attack cancer cells. This review focuses on the characteristics of PD-1, PDL-1, and CTLA-4 immune checkpoints, their mechanism of action, and their role in skin cancer. Additionally, it summarizes the ongoing clinical trials sponsored or conducted by various pharmaceutical companies and provides insights into the latest patent data.
Collapse
Affiliation(s)
- Neha Sharma
- Noida Institute of Engineering and Technology (Pharmacy Institute) 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute) 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Pallavi Rai
- Ram-Eesh Institute of Vocational and Technical Education, Greater Noida, India
| |
Collapse
|
20
|
Sabini E, Khan A, Caturegli P. Сytotoxic T lymphocyte-associated protein 4 (CTLA4) is overexpressed in a subset of prolactin- and growth hormone-secreting pituitary adenomas. Endocr Relat Cancer 2024; 31:e230196. [PMID: 37870923 PMCID: PMC11249045 DOI: 10.1530/erc-23-0196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 10/23/2023] [Indexed: 10/25/2023]
Abstract
Cytotoxic T lymphocyte-associated protein 4 (CTLA4), a negative regulator typically expressed on the surface of T lymphocytes, is targeted by immunotherapy in patients with an ever-expanding spectrum of cancers. Characterizing the expression of CTLA4 in the pituitary gland could provide additional rationale for using immune checkpoint inhibitors in pituitary adenoma patients who do not respond to conventional treatments. We assessed the expression of CTLA4 mRNA and protein in a panel of 157 human pituitary glands, 45 collected at autopsy and 112 at surgery. These specimens included 50 normal glands and 107 adenomas: 41 nonsecreting, 25 PRL-, 24 ACTH-, 11 GH-, 2 TSH-, 1 FSH-secreting, and 3 atypical. Specimens were stained for CTLA4 and adenohypophyseal hormones using RNAscope in situ hybridization, immunohistochemistry, and RNAscope Multiplex Fluorescent Assay. CTLA4 mRNA was detectable in most normal pituitary glands (48 of 50, 96%) but varied in expression, with a histological score (H-score) ranging from 0.6 to 20. The variation did not depend upon the patient's gender and age and was not significantly affected by the archival storage time. CTLA4 expression was higher (P = 0.022) in pituitary adenomas than normal glands, with the greatest levels seen in PRL- and GH-secreting adenomas (P = 0.009 and 0.023 versus normal, respectively). Eight of 25 (32%) prolactinomas and 3 of 11 (27%) GH-adenomas had an H-score greater than 20, while no differences were seen for the other types. These novel data highlight the expression of an immune checkpoint such as CTLA4 on pituitary endocrine cells, a finding that could be exploited for therapeutical applications.
Collapse
Affiliation(s)
- Elena Sabini
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
| | - Amna Khan
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
| | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
- Department of Pathology, Johns Hopkins Hospital, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
21
|
Kejamurthy P, Devi KTR. Immune checkpoint inhibitors and cancer immunotherapy by aptamers: an overview. Med Oncol 2023; 41:40. [PMID: 38158454 DOI: 10.1007/s12032-023-02267-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/22/2023] [Indexed: 01/03/2024]
Abstract
Efforts in cancer immunotherapy aim to counteract evasion mechanisms and stimulate the immune system to recognise and attack cancer cells effectively. Combination therapies that target multiple aspects of immune evasion are being investigated to enhance the overall efficacy of cancer immunotherapy. PD-1 (Programmed Cell Death Protein 1), CTLA-4 (Cytotoxic T-Lymphocyte Antigen 4), LAG-3 (Lymphocyte-Activation Gene 3), and TIM-3 (T Cell Immunoglobulin and Mucin Domain-Containing Protein3) are all immune checkpoint receptors that play crucial roles in regulating the immune response and maintaining self-tolerance often exploited by cancer cells to evade immune surveillance. Antibodies targeted against immune checkpoint inhibitors such as anti-PD-1 antibodies (e.g., pembrolizumab, nivolumab), anti-CTLA-4 antibodies (e.g., Ipilimumab), and experimental drugs targeting LAG-3 and TIM-3, aim to block these interactions and unleash the immune system's ability to recognise and destroy cancer cells. The US FDA has approved different categories of immune checkpoint inhibitors that have been utilised successfully in some patients with metastatic melanoma, renal cell carcinoma, head and neck cancers, and non-small lung cancer. Although several immune checkpoint inhibitor antibodies have been developed, they exhibited immune-related adverse effects, resulting in hypophysitis, diabetes, and neurological issues. These adverse effects of antibodies can be reduced by developing aptamer against the target. Aptamers offer several advantages over traditional antibodies, such as improved specificity, reduced immunogenicity, and flexible design for reduced adverse effects that specifically target and block protein-protein or receptor-ligand interactions involved in immune checkpoint pathways. The current study aims to review the function of particular immune checkpoint inhibitors along with developed aptamer-mediated antitumor cytotoxicity in cancer treatment.
Collapse
Affiliation(s)
- Priyatharcini Kejamurthy
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - K T Ramya Devi
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India.
| |
Collapse
|
22
|
Liu S, Jia M, Dai R. Deciphering the tumour immune microenvironment of hepatocellular carcinoma. Scand J Immunol 2023; 98:e13327. [PMID: 38441331 DOI: 10.1111/sji.13327] [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: 06/06/2023] [Revised: 08/13/2023] [Accepted: 08/28/2023] [Indexed: 03/07/2024]
Abstract
Current treatments for hepatocellular carcinoma (HCC) are less effective and prone to recurrence after surgery, so it's needed to seek new ideas for its therapy. Tumour immune microenvironment (TME) is crucial for the pathogenesis, development and metastasis of HCC. Interactions between immune cells and tumour cells significantly impact responses to immunotherapies and patient prognosis. In recent years, immunotherapies for HCC have shown promising potential, but the response rate is still unsatisfactory. Understanding their cross-talks is helpful for selecting potential therapeutic targets, predicting immunotherapy responses, determining immunotherapy efficacy, identifying prognostic markers and selecting individualized treatment options. In this paper, we reviewed the research advances on the roles of immune cells and multi-omic research associated with HCC pathogenesis and therapy, and future perspectives on TME.
Collapse
Affiliation(s)
- Sha Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Department of Pain, Daping Hospital, Army Medical University, Chongqing, China
| | - Man Jia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Rongyang Dai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| |
Collapse
|
23
|
Roy D, Gilmour C, Patnaik S, Wang LL. Combinatorial blockade for cancer immunotherapy: targeting emerging immune checkpoint receptors. Front Immunol 2023; 14:1264327. [PMID: 37928556 PMCID: PMC10620683 DOI: 10.3389/fimmu.2023.1264327] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
The differentiation, survival, and effector function of tumor-specific CD8+ cytotoxic T cells lie at the center of antitumor immunity. Due to the lack of proper costimulation and the abundant immunosuppressive mechanisms, tumor-specific T cells show a lack of persistence and exhausted and dysfunctional phenotypes. Multiple coinhibitory receptors, such as PD-1, CTLA-4, VISTA, TIGIT, TIM-3, and LAG-3, contribute to dysfunctional CTLs and failed antitumor immunity. These coinhibitory receptors are collectively called immune checkpoint receptors (ICRs). Immune checkpoint inhibitors (ICIs) targeting these ICRs have become the cornerstone for cancer immunotherapy as they have established new clinical paradigms for an expanding range of previously untreatable cancers. Given the nonredundant yet convergent molecular pathways mediated by various ICRs, combinatorial immunotherapies are being tested to bring synergistic benefits to patients. In this review, we summarize the mechanisms of several emerging ICRs, including VISTA, TIGIT, TIM-3, and LAG-3, and the preclinical and clinical data supporting combinatorial strategies to improve existing ICI therapies.
Collapse
Affiliation(s)
- Dia Roy
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Cassandra Gilmour
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Sachin Patnaik
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
24
|
Wenfan F, Manman X, Xingyuan S, Zeyong J, Jian Z, Lu D. Comparison of the profiles of first-line PD-1/PD-L1 inhibitors for advanced NSCLC lacking driver gene mutations: a systematic review and Bayesian network meta-analysis. Ther Adv Chronic Dis 2023; 14:20406223231189224. [PMID: 37841212 PMCID: PMC10568994 DOI: 10.1177/20406223231189224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 06/08/2023] [Indexed: 10/17/2023] Open
Abstract
Background Numerous first-line immune checkpoint inhibitors (ICI) were developed for patients with advanced non-small cell lung cancer (NSCLC) lacking driver gene mutations. However, this group consists of a heterogeneous patient population, for whom the optimal therapeutic choice is yet to be confirmed. Objective To identify the best first-line immunotherapy regimen for overall advanced NSCLC patients and different subgroups. Design Systematic review and Bayesian network meta-analysis (NMA). Methods We searched several databases to retrieve relevant literature. We performed Bayesian NMA for the overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and treatment-related adverse events (tr-AEs) with a grade equal or more than 3 (grade ⩾ 3 tr-AEs). Subgroup analysis was conducted according to programed death ligand 1 (PD-L1) levels, histologic type, central nervous system (CNS) metastases and tobacco use history. Results For the PD-L1 non-selective patients, sintilimab plus chemotherapy (sinti-chemo) provided the best OS [hazard ratio (HR) = 0.59, 95% confidence interval (CI):0.42-0.83]. Nivolumab plus bevacizumab plus chemotherapy (nivo-bev-chemo) was comparable to atezolizumab plus bevacizumab plus chemotherapy (atezo-bev-chemo) in prolonging PFS (HR = 0.99, 95% CI: 0.51-1.91). Atezo-bev-chemo remarkably elevated the ORR than chemotherapy (OR = 3.13, 95% CI: 1.51-6.59). Subgroup analysis showed pembrolizumab plus chemotherapy (pembro-chemo) ranked first in OS in subgroups of PD-L1 < 1%, non-squamous, no CNS metastases, with or without smoking history, and ranked second in OS in subgroups of PD-L1 ⩾ 1% and PD-L1 1-49%. Cemiplimab and sugemalimab plus chemotherapy ranked first in OS and PFS for squamous subgroup, respectively. For patients with CNS metastases, nivolumab plus ipilimumab plus chemotherapy (nivo-ipili-chemo) and camrelizumab plus chemotherapy provided the best OS and PFS, respectively. Conclusions Sinti-chemo and nivo-bev-chemo were two effective first-line regimens ranked first in OS and PFS for overall patients, respectively. Pembro-chemo was favorable for patients in subgroups of PD-L1 < 1%, PD-L1 ⩾ 1%, PD-L1 1-49%, non-squamous, no CNS metastases, with or without smoking history. Addition of bevacizumab consistently provided with favorable PFS results in patients of all PD-L1 levels. Cemiplimab was the best option in squamous subgroup and nivo-ipili-chemo in CNS metastases subgroup due to their advantages in OS.
Collapse
Affiliation(s)
- Fu Wenfan
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
- Southern Medical University, Guangzhou, Guangdong, China
- Department of Thoracic Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Xu Manman
- Department of Gynecology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shi Xingyuan
- Department of Radiotherapy, the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiang Zeyong
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhao Jian
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Dai Lu
- Department of Chest Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 78 Hengzhigang Road, Guangzhou, Guangdong 510095, China
| |
Collapse
|
25
|
Jiang D, Ma X, Zhang X, Cheng B, Wang R, Liu Y, Zhang X. New techniques: a roadmap for the development of HCC immunotherapy. Front Immunol 2023; 14:1121162. [PMID: 37426674 PMCID: PMC10323423 DOI: 10.3389/fimmu.2023.1121162] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 06/09/2023] [Indexed: 07/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide. The absence of effective early diagnostic methods and the limitations of conventional therapies have led to a growing interest in immunotherapy as a novel treatment approach for HCC. The liver serves as an immune organ and a recipient of antigens from the digestive tract, creating a distinctive immune microenvironment. Key immune cells, including Kupffer cells and cytotoxic T lymphocytes, play a crucial role in HCC development, thus offering ample research opportunities for HCC immunotherapy. The emergence of advanced technologies such as clustered regularly interspaced short palindromic repeats (CRISPR) and single-cell ribonucleic acid sequencing has introduced new biomarkers and therapeutic targets, facilitating early diagnosis and treatment of HCC. These advancements have not only propelled the progress of HCC immunotherapy based on existing studies but have also generated new ideas for clinical research on HCC therapy. Furthermore, this review analysed and summarised the combination of current therapies for HCC and the improvement of CRISPR technology for chimeric antigen receptor T cell therapy, instilling renewed hope for HCC treatment. This review comprehensively explores the advancements in immunotherapy for HCC, focusing on the use of new techniques.
Collapse
|
26
|
Medlej ZAA, Medlej W, Slaba S, Torrecillas P, Cueto A, Urbaneja A, Garrido AJ, Lugnani F. Cryoablation and Immunotherapy: An Enthralling Synergy for Cancer Treatment. Curr Oncol 2023; 30:4844-4860. [PMID: 37232823 DOI: 10.3390/curroncol30050365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/02/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
As less invasive options for surgical tumor removal, minimally invasive ablative techniques have gained popularity. Several solid tumors are being treated with cryoablation, a non-heat-based ablation technique. Cryoablation data in comparison over time demonstrates better tumor response and faster recovery. Combining cryosurgery with other cancer therapies has been explored to improve the cancer-killing process. Cryoablation with the combination of immunotherapy, results in a robust and efficient attack on the cancer cells. This article focuses on investigating the ability of cryosurgery to create a strong antitumor response when combined with immunologic agents resulting in a synergetic effect. To achieve this objective, we combined cryosurgery with immunotherapy using Nivolumab and lpilimumab. Five clinical cases of lymph node, lung cancer, bone, and lung metastasis were followed and analyzed. In this series of patients, percutaneous cryoablation and addressing immunity agents were technically feasible. In the follow-ups, there appeared to be no radiological evidence of new tumor development.
Collapse
Affiliation(s)
- Zain Al Abidine Medlej
- Agro-Food and Environmental Biosciences and Technologies Department, University of Teramo, 64100 Teramo, Italy
| | - Wassim Medlej
- Cryolebabon and Medical Devices Sarl, Beirut 1107 2020, Lebanon
| | - Sami Slaba
- Hotel Dieu de France Hospital, Saint-Joseph University, Beirut 1104 2020, Lebanon
| | | | - Antonio Cueto
- Radiology Department, Clinica Santa Elena, 29620 Madrid, Spain
| | | | | | - Franco Lugnani
- Radiology Department, Clinica Santa Elena, 29620 Madrid, Spain
| |
Collapse
|
27
|
Manso T, Kushwaha A, Abdollahi N, Duroux P, Giudicelli V, Kossida S. Mechanisms of action of monoclonal antibodies in oncology integrated in IMGT/mAb-DB. Front Immunol 2023; 14:1129323. [PMID: 37215135 PMCID: PMC10196129 DOI: 10.3389/fimmu.2023.1129323] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/07/2023] [Indexed: 05/24/2023] Open
Abstract
Background Cancer cells activate different immune checkpoint (IC) pathways in order to evade immunosurveillance. Immunotherapies involving ICs either block or stimulate these pathways and enhance the efficiency of the immune system to recognize and attack cancer cells. In this way, the development of monoclonal antibodies (mAbs) targeting ICs has significant success in cancer treatment. Recently, a systematic description of the mechanisms of action (MOA) of the mAbs has been introduced in IMGT/mAb-DB, the IMGT® database dedicated to mAbs for therapeutic applications. The characterization of these antibodies provides a comprehensive understanding of how mAbs work in cancer. Methods In depth biocuration taking advantage of the abundant literature data as well as amino acid sequence analyses from mAbs managed in IMGT/2Dstructure-DB, the IMGT® protein database, allowed to define a standardized and consistent description of the MOA of mAbs targeting immune checkpoints in cancer therapy. Results A fine description and a standardized graphical representation of the MOA of selected mAbs are integrated within IMGT/mAb-DB highlighting two main mechanisms in cancer immunotherapy, either Blocking or Agonist. In both cases, the mAbs enhance cytotoxic T lymphocyte (CTL)-mediated anti-tumor immune response (Immunostimulant effect) against tumor cells. On the one hand, mAbs targeting co-inhibitory receptors may have a functional Fc region to increase anti-tumor activity by effector properties that deplete Treg cells (Fc-effector function effect) or may have limited FcγR binding to prevent Teff cells depletion and reduce adverse events. On the other hand, agonist mAbs targeting co-stimulatory receptors may bind to FcγRs, resulting in antibody crosslinking (FcγR crosslinking effect) and substantial agonism. Conclusion In IMGT/mAb-DB, mAbs for cancer therapy are characterized by their chains, domains and sequence and by several therapeutic metadata, including their MOA. MOAs were recently included as a search criterion to query the database. IMGT® is continuing standardized work to describe the MOA of mAbs targeting additional immune checkpoints and novel molecules in cancer therapy, as well as expanding this study to other clinical domains.
Collapse
|
28
|
Wang J, Wang Y, Pan H, Zhao L, Yang X, Liang Z, Shen X, Zhang J, Yang J, Zhu Y, Xun J, Liang Y, Lin Q, Liang H, Li M, Zhu H. Chemokine Receptors CCR6 and PD1 Blocking scFv E27 Enhances Anti-EGFR CAR-T Therapeutic Efficacy in a Preclinical Model of Human Non-Small Cell Lung Carcinoma. Int J Mol Sci 2023; 24:ijms24065424. [PMID: 36982500 PMCID: PMC10056525 DOI: 10.3390/ijms24065424] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/14/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells, a therapeutic agent for solid tumors, are not completely effective due to a lack of infiltration of T cells into the tumor site and immunity caused by Programmed Death Receptor 1(PD1). Here, an epidermal growth factor receptor (EGFR) CAR-T cell was engineered to express the chemokine receptor CCR6 and secrete PD1 blocking Single-chain antibody fragment (scFv) E27 to enhance their anti-tumor effects. The findings showed that CCR6 enhanced the migration of EGFR CAR-E27-CCR6 T cells in vitro by the Transwell migration assay. When incubated with tumor cells, EGFR CAR-E27-CCR6 T cells specifically exerted potent cytotoxicity and produced high levels of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), and interferon-γ (IFN-γ). A non-small cell lung carcinoma (NSCLC) cell line-derived xenograft model was constructed by implanting modified A549 cell lines into immunodeficient NOD.PrkdcscidIl2rgem1/Smoc (NSG) mice. In comparison with traditional EGFR CAR-T cells, live imaging indicated that EGFR CAR-E27-CCR6 T cells displayed superior anti-tumor function. In addition, the histopathological examination of mouse organs showed no obvious organic damage. Our findings confirmed that PD1 blocking and CCR6 can enhance the anti-tumor function of EGFR CAR-T cells in an NSCLC xenograft model, providing an effective treatment strategy to improve the efficacy of CAR-T in NSCLC.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Yanan Wang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Hanyu Pan
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Lin Zhao
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Xinyi Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Zhiming Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Xiaoting Shen
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Jing Zhang
- School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Jinlong Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Yuqi Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Jingna Xun
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Yue Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Qinru Lin
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Huitong Liang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Min Li
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
| | - Huanzhang Zhu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200082, China
- Correspondence: ; Tel./Fax: +86-021-31246728
| |
Collapse
|
29
|
Hogan JM, Lee PS, Wong SC, West SM, Morishige WH, Bee C, Tapia GC, Rajpal A, Strop P, Dollinger G. Residue-Level Characterization of Antibody Binding Epitopes Using Carbene Chemical Footprinting. Anal Chem 2023; 95:3922-3931. [PMID: 36791402 DOI: 10.1021/acs.analchem.2c03091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Characterization of antibody binding epitopes is an important factor in therapeutic drug discovery, as the binding site determines and drives antibody pharmacology and pharmacokinetics. Here, we present a novel application of carbene chemical footprinting with mass spectrometry for identification of antibody binding epitopes at the single-residue level. Two different photoactivated diazirine reagents provide complementary labeling information allowing structural refinement of the antibody binding interface. We applied this technique to map the epitopes of multiple MICA and CTLA-4 antibodies and validated the findings with X-ray crystallography and yeast surface display epitope mapping. The characterized epitopes were used to understand biolayer interferometry-derived competitive binding results at the structural level. We show that carbene footprinting provides fast and high-resolution epitope information critical in the antibody selection process and enables mechanistic understanding of function to accelerate the drug discovery process.
Collapse
Affiliation(s)
- Jason M Hogan
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Peter S Lee
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Susan C Wong
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Sean M West
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Winse H Morishige
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Christine Bee
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Gamze Camdere Tapia
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Arvind Rajpal
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Pavel Strop
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| | - Gavin Dollinger
- Discovery Biotherapeutics, Bristol Myers Squibb, 700 Bay Road, Redwood City, California 94063, United States
| |
Collapse
|
30
|
Bouqdayr M, Abbad A, Baba H, Saih A, Wakrim L, Kettani A. Computational analysis of structural and functional evaluation of the deleterious missense variants in the human CTLA4 gene. J Biomol Struct Dyn 2023; 41:14179-14196. [PMID: 36764830 DOI: 10.1080/07391102.2023.2178509] [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: 11/18/2022] [Accepted: 02/04/2023] [Indexed: 02/12/2023]
Abstract
CTLA-4 is an immune checkpoint receptor that negatively regulates the T-cell function expressed after T-cell activation to break the immune response. The current study predicted the genomic analysis to explore the functional variations of missense SNPs in the human CTLA4 gene using PolyPhen2, SIFT, PANTHER, PROVEAN, Fathmm, Mutation Assessor, PhD-SNP, SNPs&GO, SNAP2, and MutPred2. Phylogenetic conservation protein was predicted by ConSurf. Protein structural analysis was carried out by I-Mutant3, MUpro, iStable2, PremPS, and ERIS servers. Molecular dynamics trajectory analysis (RMSD, RMSF, Rg, SASA, H-bonds, and PCA) was performed to analyze the dynamic behavior of native and mutant CTLA-4 at the atomic level. Our in-silico analysis suggested that C58S, G118R, P137Q, P137R, P137L, P138T, and G146L variants were predicted to be the most deleterious missense variants and highly conserved residues. Moreover, the molecular dynamics analysis proposed a decrease in the protein stability and compactness with the P137R and P138T highlighting the impact of these variants on the function of the CTLA-4 protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Meryem Bouqdayr
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Anass Abbad
- Medical Virology and BSL-3 Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Hanâ Baba
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Asmae Saih
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Lahcen Wakrim
- Virology Unit, Immunovirology Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Anass Kettani
- Laboratory of Biology and Health, Faculty of Sciences Ben M'sick, Hassan II University of Casablanca, Casablanca, Morocco
| |
Collapse
|
31
|
Guan J, Liu H, Chai Y, Yu J, Yao J, Wang J, Pan Z, Zhang J, Zhou Y, Liu H, Yao S, Qi J, Feng H, Gao GF, Wang Q, Shi Y, Tan S. Characterization of the high-affinity anti-CTLA-4 monoclonal antibody JS007 for immune checkpoint therapy of cancer. MAbs 2023; 15:2153409. [PMID: 36511654 PMCID: PMC9754112 DOI: 10.1080/19420862.2022.2153409] [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] [Indexed: 12/15/2022] Open
Abstract
Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is a critical inhibitory checkpoint molecule, and monoclonal antibodies (mAbs) targeting CTLA-4 that restore anti-tumor T cell immunity have achieved clinical success. Here, we report a humanized IgG1 mAb, namely JS007, with high binding affinity to CTLA-4. JS007 shows superior binding affinity and T-cell activating efficiency over ipilimumab. Moreover, it demonstrates substantial in vivo tumor suppression efficacy at low doses. The crystal structure of JS007/CTLA-4 complex (PDB: 8HIT) shows JS007 adopts a heavy-chain-dominant binding mode, and mainly contacts the BC loop, DE loop and FG loop of CTLA-4. Notably, two Tyr residues (VH-Y100 and VL-Y32) from the complementarity-determining region loops insert into the two cavities formed by the residues from the loops of CTLA-4, which may contribute to the stabilization of the binding. Comparative analysis with other anti-CTLA-4 mAbs indicates that the double "wedge-into-hole" binding mode is unique for JS007 and may be responsible for the high-affinity binding to CTLA-4. These findings have provided an important molecular understanding of the high-affinity CTLA-4 blockade mAbs and shed light on future development of agents targeting CTLA-4.
Collapse
Affiliation(s)
- Jiawei Guan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Hongchuan Liu
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Yan Chai
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China
| | - Jie Yu
- Pilot National Laboratory for Marine Science and Technology (Qingdao), Shandong, China
| | - Jian Yao
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jing Wang
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Zhiwei Pan
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jing Zhang
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Yuehua Zhou
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Hui Liu
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Sheng Yao
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - Jianxun Qi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China
| | - Hui Feng
- Department of Antibody Discovery and Engineering, Shanghai Junshi Biosciences Co Ltd, Shanghai, China
| | - George F. Gao
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Qihui Wang
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China,CONTACT Qihui Wang
| | - Yi Shi
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Shuguang Tan
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Dang X, Guelen L, Lutje Hulsik D, Ermakov G, Hsieh EJ, Kreijtz J, Stammen-Vogelzangs J, Lodewijks I, Bertens A, Bramer A, Guadagnoli M, Nazabal A, van Elsas A, Fischmann T, Juan V, Beebe A, Beaumont M, van Eenennaam H. Epitope mapping of monoclonal antibodies: a comprehensive comparison of different technologies. MAbs 2023; 15:2285285. [PMID: 38010385 PMCID: PMC10730160 DOI: 10.1080/19420862.2023.2285285] [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: 11/02/2022] [Accepted: 11/15/2023] [Indexed: 11/29/2023] Open
Abstract
Monoclonal antibodies have become an important class of therapeutics in the last 30 years. Because the mechanism of action of therapeutic antibodies is intimately linked to their binding epitopes, identification of the epitope of an antibody to the antigen plays a central role during antibody drug development. The gold standard of epitope mapping, X-ray crystallography, requires a high degree of proficiency with no guarantee of success. Here, we evaluated six widely used alternative methods for epitope identification (peptide array, alanine scan, domain exchange, hydrogen-deuterium exchange, chemical cross-linking, and hydroxyl radical footprinting) in five antibody-antigen combinations (pembrolizumab+PD1, nivolumab+PD1, ipilimumab+CTLA4, tremelimumab+CTLA4, and MK-5890+CD27). The advantages and disadvantages of each technique are demonstrated by our data and practical advice on when and how to apply specific epitope mapping techniques during the drug development process is provided. Our results suggest chemical cross-linking most accurately identifies the epitope as defined by crystallography.
Collapse
Affiliation(s)
- Xibei Dang
- Pharmacokinetics, Merck & Co. Inc, Kenilworth, NJ, USA
| | - Lars Guelen
- Research, Aduro Biotech Europe, Oss, The Netherlands
| | | | | | | | - Joost Kreijtz
- Research, Aduro Biotech Europe, Oss, The Netherlands
| | | | | | | | - Arne Bramer
- Research, Aduro Biotech Europe, Oss, The Netherlands
| | | | | | | | | | - Veronica Juan
- Pharmacokinetics, Merck & Co. Inc, Kenilworth, NJ, USA
| | - Amy Beebe
- Pharmacokinetics, Merck & Co. Inc, Kenilworth, NJ, USA
| | | | | |
Collapse
|
33
|
Chen Z, Meng L, Zhang J, Zhang X. Progress in the cryoablation and cryoimmunotherapy for tumor. Front Immunol 2023; 14:1094009. [PMID: 36761748 PMCID: PMC9907027 DOI: 10.3389/fimmu.2023.1094009] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
With the rapid advancement of imaging equipment and minimally invasive technology, cryoablation technology is being used more frequently in minimally invasive treatment of tumors, primarily for patients with early tumors who voluntarily consent to ablation as well as those with advanced tumors that cannot be surgically removed or cannot be tolerated. Cryoablation is more effective and secure for target lesions than other thermal ablation methods like microwave and radiofrequency ablation (RFA). The study also discovered that cryoablation, in addition to causing tumor tissue necrosis and apoptosis, can facilitate the release of tumor-derived autoantigens into the bloodstream and activate the host immune system to elicit beneficial anti-tumor immunological responses against primary. This may result in regression of the primary tumor and distant metastasis. The additional effect called " Accompanying effects ". It is the basis of combined ablation and immunotherapy for tumor. At present, there is a lot of research on the mechanism of immune response induced by cryoablation. Trying to solve the question: how positively induce immune response. In this review, we focus on: 1. the immune effects induced by cryoablation. 2. the effect and mechanism of tumor immunotherapy combined with cryoablation. 3.The clinical research of this combination therapy in the treatment of tumors.
Collapse
Affiliation(s)
- Zenan Chen
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Liangliang Meng
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China.,Department of Radiology, Chinese People's Armed Police (PAP) Force Hospital of Beijing, Beijing, China
| | - Jing Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Xiao Zhang
- Department of Radiology, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| |
Collapse
|
34
|
George A, Varghese J, Padinharayil H. Potential of Biotechnology in Cancer Management. NOVEL TECHNOLOGIES IN BIOSYSTEMS, BIOMEDICAL & DRUG DELIVERY 2023:9-44. [DOI: 10.1007/978-981-99-5281-6_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Guerrisi A, Falcone I, Valenti F, Rao M, Gallo E, Ungania S, Maccallini MT, Fanciulli M, Frascione P, Morrone A, Caterino M. Artificial Intelligence and Advanced Melanoma: Treatment Management Implications. Cells 2022; 11:cells11243965. [PMID: 36552729 PMCID: PMC9777238 DOI: 10.3390/cells11243965] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Artificial intelligence (AI), a field of research in which computers are applied to mimic humans, is continuously expanding and influencing many aspects of our lives. From electric cars to search motors, AI helps us manage our daily lives by simplifying functions and activities that would be more complex otherwise. Even in the medical field, and specifically in oncology, many studies in recent years have highlighted the possible helping role that AI could play in clinical and therapeutic patient management. In specific contexts, clinical decisions are supported by "intelligent" machines and the development of specific softwares that assist the specialist in the management of the oncology patient. Melanoma, a highly heterogeneous disease influenced by several genetic and environmental factors, to date is still difficult to manage clinically in its advanced stages. Therapies often fail, due to the establishment of intrinsic or secondary resistance, making clinical decisions complex. In this sense, although much work still needs to be conducted, numerous evidence shows that AI (through the processing of large available data) could positively influence the management of the patient with advanced melanoma, helping the clinician in the most favorable therapeutic choice and avoiding unnecessary treatments that are sure to fail. In this review, the most recent applications of AI in melanoma will be described, focusing especially on the possible finding of this field in the management of drug treatments.
Collapse
Affiliation(s)
- Antonino Guerrisi
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, 00144 Rome, Italy
| | - Italia Falcone
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence:
| | - Fabio Valenti
- UOC Oncological Translational Research, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Marco Rao
- Enea-FSN-TECFIS-APAM, C.R. Frascati, via Enrico Fermi, 45, 00146 Rome, Italy
| | - Enzo Gallo
- Pathology Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Sara Ungania
- Medical Physics and Expert Systems Laboratory, Department of Research and Advanced Technologies, IRCCS-Regina Elena Institute, 00144 Rome, Italy
| | - Maria Teresa Maccallini
- Departement of Clinical and Molecular Medicine, Università La Sapienza di Roma, 00185 Rome, Italy
| | - Maurizio Fanciulli
- SAFU, Department of Research, Advanced Diagnostics, and Technological Innovation, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Pasquale Frascione
- Oncologic and Preventative Dermatology, IFO-San Gallicano Dermatological Institute-IRCCS, 00144 Rome, Italy
| | - Aldo Morrone
- Scientific Direction, San Gallicano Dermatological Institute IRCCS, 00144 Rome, Italy
| | - Mauro Caterino
- Radiology and Diagnostic Imaging Unit, Department of Clinical and Dermatological Research, San Gallicano Dermatological Institute IRCCS, 00144 Rome, Italy
| |
Collapse
|
36
|
Morano NC, Smith RS, Danelon V, Schreiner R, Patel U, Herrera NG, Smith C, Olson SM, Laerke MK, Celikgil A, Garforth SJ, Garrett-Thomson SC, Lee FS, Hempstead BL, Almo SC. Human immunomodulatory ligand B7-1 mediates synaptic remodeling via the p75 neurotrophin receptor. J Clin Invest 2022; 132:e157002. [PMID: 36107635 PMCID: PMC9663165 DOI: 10.1172/jci157002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 09/13/2022] [Indexed: 12/30/2023] Open
Abstract
Cell surface receptors, ligands, and adhesion molecules underlie development, circuit formation, and synaptic function of the central nervous system and represent important therapeutic targets for many neuropathologies. The functional contributions of interactions between cell surface proteins of neurons and nonneuronal cells have not been fully addressed. Using an unbiased protein-protein interaction screen, we showed that the human immunomodulatory ligand B7-1 (hB7-1) interacts with the p75 neurotrophin receptor (p75NTR) and that the B7-1:p75NTR interaction is a recent evolutionary adaptation present in humans and other primates, but absent in mice, rats, and other lower mammals. The surface of hB7-1 that engages p75NTR overlaps with the hB7-1 surface involved in CTLA-4/CD28 recognition, and these molecules directly compete for binding to p75NTR. Soluble or membrane-bound hB7-1 altered dendritic morphology of cultured hippocampal neurons, with loss of the postsynaptic protein PSD95 in a p75NTR-dependent manner. Abatacept, an FDA-approved therapeutic (CTLA-4-hFc fusion) inhibited these processes. In vivo injection of hB7-1 into the murine subiculum, a hippocampal region affected in Alzheimer's disease, resulted in p75NTR-dependent pruning of dendritic spines. Here, we report the biochemical interaction between B7-1 and p75NTR, describe biological effects on neuronal morphology, and identify a therapeutic opportunity for treatment of neuroinflammatory diseases.
Collapse
Affiliation(s)
- Nicholas C. Morano
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, USA
| | - Roshelle S. Smith
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Victor Danelon
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Ryan Schreiner
- Division of Regenerative Medicine, Hartman Institute for Therapeutic Organ Regeneration, Ansary Stem Cell Institute, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Uttsav Patel
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Natalia G. Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Carla Smith
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Steven M. Olson
- Department of Computer Science, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Michelle K. Laerke
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Alev Celikgil
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | - Scott J. Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| | | | - Francis S. Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, New York, USA
| | - Barbara L. Hempstead
- Department of Medicine, Weill Cornell Graduate School of Medical Sciences, New York, New York, USA
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, New York, New York, USA
| |
Collapse
|
37
|
Fei X, Liu S, Liu P, Wang X, Zhu C, Hou J, Cai J, Pan Y. Identification and validation of a potential key gene SGOL1 for poor prognosis in hepatocellular carcinoma based on a bioinformatics approach. Front Oncol 2022; 12:1043161. [PMID: 36439418 PMCID: PMC9683735 DOI: 10.3389/fonc.2022.1043161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/21/2022] [Indexed: 08/15/2023] Open
Abstract
OBJECTIVE Hepatocellular carcinoma (HCC) is one of the most prevalent types of cancer worldwide. Shugoshin 1 (SGOL1) plays a crucial role in cell mitosis and its aberrant expression level in human tumors has shown to promote chromosomal instability (CIN) and accelerate tumor growth. SGOL1 expression level in HCC cells and tissues, whether it has an influence on HCC patients' prognosis, and its mechanism of action have not yet been studied. METHODS We carried out the bioinformatics analysis of SGOL1 expression level and survival analysis in 8 different malignancies, including HCC. In addition, we analyzed SGOL1 expression level in HCC tissues, as well as HCC patients' clinical features, enrichment analysis of SGOL1 function and mechanism of action in HCC and tumor immune cells. The effects of SGOL1 expression level and cell viability on HCC were confirmed by in vitro cytological assays. RESULTS It was found that SGOL1 mRNA expression level was significantly higher in several tumor tissues, including HCC, than in corresponding normal tissues, and the elevated SGOL1 expression level was strongly associated with HCC patients' poor prognosis. It was also revealed that SGOL1 expression level in HCC tissue was positively correlated with disease stage, tumor grade, and tumor size, and the results of multivariate logistic regression analysis showed that SGOL1 was one of the independent influential factors of the prognosis of HCC. Enrichment analysis revealed that SGOL1 expression level in HCC tissue was mainly associated with tumor proliferation, cell cycle, and other factors. The results of the immune infiltration analysis indicated that SGOL1 expression level was associated with immune cell infiltration and immune checkpoints in HCC. In vitro experiments demonstrated the high SGOL1 expression level in HCC tissues and cells, and silencing of SGOL1 resulted in altered cell cycle markers and decreased proliferation, invasion, and migration of HCC cells. CONCLUSION The findings revealed that SGOL1 is highly expressed in HCC tissues, it is a biomarker of a poor prognosis, which may be related to immune cell infiltration in HCC, and may enhance the proliferation, invasion, and migration of HCC cells. The results may provide new insights into targeted treatment of HCC and improve HCC patients' prognosis.
Collapse
Affiliation(s)
- Xiaobin Fei
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Songbai Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Peng Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xing Wang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Changhao Zhu
- Department of Hepatobiliary Surgery, Guizhou Cancer Hospital, Guiyang, China
| | - Junyi Hou
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Junzhe Cai
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yaozhen Pan
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
38
|
Lee DY, Im E, Yoon D, Lee YS, Kim GS, Kim D, Kim SH. Pivotal role of PD-1/PD-L1 immune checkpoints in immune escape and cancer progression: Their interplay with platelets and FOXP3+Tregs related molecules, clinical implications and combinational potential with phytochemicals. Semin Cancer Biol 2022; 86:1033-1057. [PMID: 33301862 DOI: 10.1016/j.semcancer.2020.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/25/2020] [Accepted: 12/01/2020] [Indexed: 01/27/2023]
Abstract
Immune checkpoint proteins including programmed cell death protein 1 (PD-1), its ligand PD-L1 and cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) are involved in proliferation, angiogenesis, metastasis, chemoresistance via immune escape and immune tolerance by disturbing cytotoxic T cell activation. Though many clinical trials have been completed in several cancers by using immune checkpoint inhibitors alone or in combination with other agents to date, recently multi-target therapy is considered more attractive than monotherapy, since immune checkpoint proteins work with other components such as surrounding blood vessels, dendritic cells, fibroblasts, macrophages, platelets and extracellular matrix within tumor microenvironment. Thus, in the current review, we look back on research history of immune checkpoint proteins and discuss their associations with platelets or tumor cell induced platelet aggregation (TCIPA) and FOXP3+ regulatory T cells (Tregs) related molecules involved in immune evasion and tumor progression, clinical implications of completed trial results and signaling networks by phytochemicals for combination therapy with immune checkpoint inhibitors and suggest future research perspectives.
Collapse
Affiliation(s)
- Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Eunji Im
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Dahye Yoon
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Young-Seob Lee
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Geum-Soog Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Donghwi Kim
- Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA, Eumseong, 27709, Republic of Korea
| | - Sung-Hoon Kim
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
39
|
Ren X, Liu J, Wang R, Liu X, Ma X, Lu Z, Hu Z, Zheng M, Ma J, Li J. Exploring the oncogenic roles of LINC00857 in pan-cancer. Front Pharmacol 2022; 13:996686. [PMID: 36160408 PMCID: PMC9498830 DOI: 10.3389/fphar.2022.996686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/12/2022] [Indexed: 11/30/2022] Open
Abstract
Although aberrant LINC00857 expression may play a key role in oncogenesis, no research has analyzed the pan-cancer oncogenic roles of LINC00857, particularly in tumor immunology. Here, we integrated data from several databases to analyze the characteristics of LINC00857 in pan-cancer. We found that LINC00857 was overexpressed and correlated with a poor prognosis in a variety of cancers. Furthermore, high-expression of LINC00857 was negatively associated with immune cell infiltration and immune checkpoint gene expression. Notably, LINC00857 expression was negatively related to microsatellite instability and tumor mutation burden in colorectal cancer, implying poor reaction to immunotherapy when LINC00857 was highly expressed. Targeting LINC00857 could dramatically impair the proliferative ability of colorectal cancer cells. After RNA-sequencing in HCT116 cells, gene set enrichment analysis showed that LINC00857 may accelerate cancer progression by inhibiting the ferroptosis pathway and promoting glycolipid metabolism in colorectal cancer. Screening by weighted gene co-expression network analysis determined PIWIL4 as a target of LINC00857, which also performed an immunosuppressive role in colorectal cancer. Based on the structure of PIWIL4, a number of small molecule drugs were screened out by virtual screening and sensitivity analysis. In summary, LINC00857 expression was closely correlated with an immunosuppressive microenvironment and may be a novel diagnostic and prognostic biomarker for diverse cancers. The LINC00857/PIWIL4 axis may be predictive biomarkers for immunotherapy and valuable molecular targets for malignant tumors.
Collapse
Affiliation(s)
- Xiaomin Ren
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Jing Liu
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Rui Wang
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Xinling Liu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Zhong Lu
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Zhenbo Hu
- Department of Hematology, Laboratory for Stem Cell and Regenerative Medicine, Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Mingzhu Zheng
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Jingang Ma
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
| | - Jiaqiu Li
- Department of Oncology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, Shandong, China
- *Correspondence: Jiaqiu Li,
| |
Collapse
|
40
|
Deng X, Chen K, Ren J, Zeng J, Zhang Q, Li T, Tang Q, Zhu J. A B7-CD28 Family-Based Signature Demonstrates Significantly Different Prognosis and Immunological Characteristics in Diffuse Gliomas. Front Mol Biosci 2022; 9:849723. [PMID: 35928223 PMCID: PMC9344576 DOI: 10.3389/fmolb.2022.849723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
The B7-CD28 gene family plays a crucial role in modulating immune functions and has served as potential targets for immunotherapeutic strategies. Therefore, we systematically analyzed B7-CD28 family gene expression profiles and constructed a B7-CD28 family-based prognostic signature to predict survival and immune host status in diffuse gliomas. The TCGA dataset was used as a training cohort, and three CGGA datasets (mRNAseq_325, mRNAseq_693 and mRNA-array) were employed as validation cohorts to intensify the findings that we have revealed in TCGA dataset. Ultimately, we developed a B7-CD28 family-based signature that consisted of CD276, CD274, PDCD1LG2 and CD80 using LASSO Cox analysis. This gene signature was validated to have significant prognostic value, and could be used as a biomarker to distinguish pathological grade and IDH mutation status in diffuse glioma. Additionally, we found that the gene signature was significantly related to intensity of immune response and immune cell population, as well as several other important immune checkpoint genes, holding a great potential to be a predictive immune marker for immunotherapy and tumor microenvironment. Finally, a B7-CD28 family-based nomogram was established to predict patient life expectancy contributing to facilitate personalizing therapy for tumor sufferers. In summary, this is the first mathematical model based on this gene family with the aim of providing novel insights into immunotherapy for diffuse glioma.
Collapse
|
41
|
|
42
|
Singh S, Kumar K, Panda M, Srivastava A, Mishra A, Prajapati VK. High-throughput virtual screening of small-molecule inhibitors targeting immune cell checkpoints to discover new immunotherapeutics for human diseases. Mol Divers 2022; 27:729-751. [PMID: 35633442 DOI: 10.1007/s11030-022-10452-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/22/2022] [Indexed: 12/28/2022]
Abstract
Immunotherapy is widely used to treat various cancers, and the drugs used are called immune checkpoint (ICP) inhibitors. Overexpression of immune cell checkpoints is reported for other human diseases such as acute infections (malaria), chronic viral infection (HIV, hepatitis B virus, TB infections), allergy, asthma, neurodegeneration, and autoimmune diseases. Some mAbs (monoclonal antibodies) are available against ICPs, but they have side effects. Small molecule seems to be safer in comparison with mAbs. Three independent small-molecule inhibitor libraries consisting of 9466 compounds were screened against seven immune cell checkpoints by applying high-throughput virtual screening approach. A total of 13 ICP inhibitors were finalized based on docking, MM-GBSA scores, and ADME properties. Six compounds were selected for MD simulation, and then, rutin hydrate (targeting all seven immune cell checkpoints), amikacin hydrate (targeting six), and 6-hydroxyluteolin (targeting three) were found to be the best immune cell checkpoint inhibitors. These three potential inhibitors have shown the potential to activate human immune cells and thus may control the spread of human lifestyle or infectious diseases. Proposed inhibitors warrant the in vitro and in vivo validation to develop it as an immunotherapeutic.
Collapse
Affiliation(s)
- Satyendra Singh
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | - Ketan Kumar
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | - Mamta Panda
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India
| | | | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, 342011, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
43
|
Kasichayanula S, Mandlekar S, Shivva V, Patel M, Girish S. Evolution of Preclinical Characterization and Insights into Clinical Pharmacology of Checkpoint Inhibitors Approved for Cancer Immunotherapy. Clin Transl Sci 2022; 15:1818-1837. [PMID: 35588531 PMCID: PMC9372426 DOI: 10.1111/cts.13312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer immunotherapy has significantly advanced the treatment paradigm in oncology, with approvals of immuno‐oncology agents for over 16 indications, many of them first line. Checkpoint inhibitors (CPIs) are recognized as an essential backbone for a successful anticancer therapy regimen. This review focuses on the US Food and Drug Administration (FDA) regulatory approvals of major CPIs and the evolution of translational advances since their first approval close to a decade ago. In addition, critical preclinical and clinical pharmacology considerations, an overview of the pharmacokinetic and dose/regimen aspects, and a discussion of the future of CPI translational and clinical pharmacology as combination therapy becomes a mainstay of industrial immunotherapy development and in clinical practice are also discussed.
Collapse
Affiliation(s)
| | | | - Vittal Shivva
- Genentech, 1 DNA Way, South San Francisco, 94080, CA
| | - Maulik Patel
- AbbVie Inc., 1000 Gateway Blvd, South San Francisco, 94080, CA
| | - Sandhya Girish
- Gilead Sciences, 310 Lakeside Drive, Foster City, 94404, CA
| |
Collapse
|
44
|
Fongaro B, Cian V, Gabaldo F, De Paoli G, Miolo G, Polverino de Laureto P. Managing antibody stability: effects of stressors on Ipilimumab from the commercial formulation to diluted solutions. Eur J Pharm Biopharm 2022; 176:54-74. [PMID: 35595030 DOI: 10.1016/j.ejpb.2022.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/19/2022]
Abstract
The stability of the monoclonal antibody Ipilimumab, the active ingredient of Yervoy®, used for the treatment of different types of cancer, has been investigated. Shaking/temperature, light exposure and dilution, protein drug renowned stressors, were applied on a 30-45-day series of experiments to observe the physicochemical and biological behavior of the molecule. Ipilimumab demonstrated stability under shaking and heat up to 45 days, without any unfolding during the induced combined stressors. Under artificial sunlight, the mAb showed to be sensitive even under the minimum dose tested (720 kJ/m2) with formation of aggregates, particularly when diluted in glucose solution. The light-induced soluble aggregates were higher in the case of diluted samples irradiated with much higher light doses (10460 kJ/m2). The aggregation of Ipilimumab took place also by irradiating the non-diluted formulation, indicating that the excipients did not protect completely the drug from photodegradation. Amino acid oxidation and deamidation were found. Anyway, after irradiation with both light doses, soluble Ipilimumab maintained its typical β-sheets structure, and the tertiary structure was nearly maintained compared to the dark. As an additional stressor test, the effect of dilution on the formulation was monitored by using a saline solution (1 mg/mL Ipilimumab) applied during hospital infusion. After two days from dilution, the protein exhibited aggregation and chemical modifications including oxidation and deamidation. When stability conditions were compromised, the viability of human cell lines treated with the stressed formulation slight decreased suggesting low potential biological toxicity of the modified mAb. As this study has demonstrated the susceptibility of Ipilimumab to light, specific solutions, and excipients as well as the use of safe light in manufacturing, handling, and storage of this drug should be promoted. Moreover, the use of proper primary and secondary packaging should be indicated to avoid the detrimental effect of light on the mAb structure and efficacy. A detailed understanding of Ipilimumab physicochemical properties, integrity, and stability could assure the best storage and manipulation conditions for its safe and successful application in cancer therapy.
Collapse
Affiliation(s)
- Benedetta Fongaro
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo, 5, 35131 Padova, Italy
| | - Valentina Cian
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo, 5, 35131 Padova, Italy
| | - Francesca Gabaldo
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo, 5, 35131 Padova, Italy
| | - Giorgia De Paoli
- Molecular and Clinical Medicine, School of Medicine, University of Dundee Nethergate, Dundee, Scotland DD1 4HN, UK
| | - Giorgia Miolo
- Department of Pharmaceutical and Pharmacological Sciences, Via Marzolo, 5, 35131 Padova, Italy.
| | | |
Collapse
|
45
|
Ding P, Ma Z, Liu D, Pan M, Li H, Feng Y, Zhang Y, Shao C, Jiang M, Lu D, Han J, Wang J, Yan X. Lysine Acetylation/Deacetylation Modification of Immune-Related Molecules in Cancer Immunotherapy. Front Immunol 2022; 13:865975. [PMID: 35585975 PMCID: PMC9108232 DOI: 10.3389/fimmu.2022.865975] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
As major post-translational modifications (PTMs), acetylation and deacetylation are significant factors in signal transmission and cellular metabolism, and are modulated by a dynamic process via two pivotal categories of enzymes, histone acetyltransferases (HATs) and histone deacetylases (HDACs). In previous studies, dysregulation of lysine acetylation and deacetylation has been reported to be associated with the genesis and development of malignancy. Scientists have recently explored acetylation/deacetylation patterns and prospective cancer therapy techniques, and the FDA has approved four HDAC inhibitors (HDACi) to be used in clinical treatment. In the present review, the most recent developments in the area of lysine acetylation/deacetylation alteration in cancer immunotherapy were investigated. Firstly, a brief explanation of the acetylation/deacetylation process and relevant indispensable enzymes that participate therein is provided. Subsequently, a multitude of specific immune-related molecules involved in the lysine acetylation/deacetylation process are listed in the context of cancer, in addition to several therapeutic strategies associated with lysine acetylation/deacetylation modification in cancer immunotherapy. Finally, a number of prospective research fields related to cancer immunotherapy concepts are offered with detailed analysis. Overall, the present review may provide a reference for researchers in the relevant field of study, with the aim of being instructive and meaningful to further research as well as the selection of potential targets and effective measures for future cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Zhiqiang Ma
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Dong Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Huizi Li
- Department of Outpatient, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Yingtong Feng
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
| | - Menglong Jiang
- Department of Thoracic Surgery, 1st Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| | - Jinliang Wang
- Department of Medical Oncology, Senior Department of Oncology, Chinese People'’s Liberation Army of China (PLA) General Hospital, The Fifth Medical Center, Beijing, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Air Force Military Medical University, Xi’an, China
- *Correspondence: Jing Han, ; Jinliang Wang, ; Xiaolong Yan,
| |
Collapse
|
46
|
Bari S, Muzaffar J, Eroglu Z. Combination targeted and immune therapy in the treatment of advanced melanoma: a valid treatment option for patients? Ther Adv Med Oncol 2022; 14:17588359221090306. [PMID: 35478991 PMCID: PMC9036333 DOI: 10.1177/17588359221090306] [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: 09/27/2021] [Accepted: 03/10/2022] [Indexed: 11/16/2022] Open
Abstract
Melanomas harboring an activating BRAFV600 mutation account for 50% of all advanced melanomas. The approval of BRAF-targeted therapy revolutionized treatment of these patients with achievement of impressive responses. However, development of resistance to these drugs is a significant problem, and as such, duration of response remains low, with median progression free survival of around 11–15 months. Immune checkpoint blockers exploit the immune system to eradicate cancer and can produce durable disease control that results in long-term, treatment-free survival in some patients. These drugs have shown very impressive survival in patients with BRAF-mutated melanoma. Thus, there is a need to continue to utilize emerging data to achieve long-term disease control for patients with advanced melanoma. Combining targeted therapy with immune therapy may be one possible way to achieve this goal. In this review, the mechanisms of action of these two pathways, including the mechanistic basis of this combination, are summarized, along with results of completed and ongoing trials in triple therapy.
Collapse
Affiliation(s)
- Shahla Bari
- Department of Cutaneous Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Jameel Muzaffar
- Department of Head and Neck Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Zeynep Eroglu
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
47
|
Cheng Z, Du Y, Yu L, Yuan Z, Tian J. Application of Noninvasive Imaging to Combined Immune Checkpoint Inhibitors for Breast Cancer: Facts and Future. Mol Imaging Biol 2022; 24:264-279. [PMID: 35102468 DOI: 10.1007/s11307-021-01688-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/13/2021] [Accepted: 11/24/2021] [Indexed: 12/19/2022]
Abstract
With the application of mono-immunotherapy in cancer, particularly immune checkpoint inhibitors, improved outcomes have been achieved. However, there are several limitations to immunotherapy, such as a poor response to the drugs, immune resistance, and immune-related adverse events. In recent years, studies of preclinical animal models and clinical trials have demonstrated that immune checkpoint inhibitors for breast cancer can significantly prolong the overall survival and quality of patients' lives. Meanwhile, combined immune checkpoint inhibitor treatment has attracted researchers' attention and showed great potential in the comprehensive treatment of breast cancer patients. Additionally, noninvasive imaging enables physicians to predict response to combined immunotherapeutic drugs, achieve treatment efficacy, and lead to better clinical management. Herein, we review the background of combined immune checkpoint inhibitor therapy and summarize its targeted imaging as well as progress in noninvasive imaging aimed at evaluating therapeutic outcomes. Finally, we describe several factors that may influence the outcome of this combined immunotherapy, the future direction of medical imaging, and the potential application of artificial intelligence in breast cancer. With further development of noninvasive imaging for the guidance of combined immune checkpoint inhibitors, cures for this disease may be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
| | - Leyi Yu
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing, 100050, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Key Laboratory of Molecular Imaging, The State Key Laboratory of Management and Control for Complex System, Institute of Automation, Chinese Academy of Sciences, BeijingBeijing, 100190, China.
- University of Chinese Academy of Sciences, Beijing, 100080, China.
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine Science and Engineering, Beihang University, Beijing, 100191, China.
- School of Life Science and Technology, Xidian University, Xi'an, Shaanxi, 710071, China.
| |
Collapse
|
48
|
Hong MMY, Maleki Vareki S. Addressing the Elephant in the Immunotherapy Room: Effector T-Cell Priming versus Depletion of Regulatory T-Cells by Anti-CTLA-4 Therapy. Cancers (Basel) 2022; 14:1580. [PMID: 35326731 PMCID: PMC8946681 DOI: 10.3390/cancers14061580] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Cytotoxic T-lymphocyte Associated Protein 4 (CTLA-4) is an immune checkpoint molecule highly expressed on regulatory T-cells (Tregs) that can inhibit the activation of effector T-cells. Anti-CTLA-4 therapy can confer long-lasting clinical benefits in cancer patients as a single agent or in combination with other immunotherapy agents. However, patient response rates to anti-CTLA-4 are relatively low, and a high percentage of patients experience severe immune-related adverse events. Clinical use of anti-CTLA-4 has regained interest in recent years; however, the mechanism(s) of anti-CTLA-4 is not well understood. Although activating T-cells is regarded as the primary anti-tumor mechanism of anti-CTLA-4 therapies, mounting evidence in the literature suggests targeting intra-tumoral Tregs as the primary mechanism of action of these agents. Tregs in the tumor microenvironment can suppress the host anti-tumor immune responses through several cell contact-dependent and -independent mechanisms. Anti-CTLA-4 therapy can enhance the priming of T-cells by blockading CD80/86-CTLA-4 interactions or depleting Tregs through antibody-dependent cellular cytotoxicity and phagocytosis. This review will discuss proposed fundamental mechanisms of anti-CTLA-4 therapy, novel uses of anti-CTLA-4 in cancer treatment and approaches to improve the therapeutic efficacy of anti-CTLA-4.
Collapse
Affiliation(s)
- Megan M Y Hong
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 3K7, Canada;
| | - Saman Maleki Vareki
- Department of Pathology and Laboratory Medicine, University of Western Ontario, London, ON N6A 3K7, Canada;
- London Regional Cancer Program, Lawson Health Research Institute, London, ON N6A 5W9, Canada
- Department of Oncology, University of Western Ontario, London, ON N6A 3K7, Canada
- Department of Medical Biophysics, University of Western Ontario, London, ON N6A 3K7, Canada
| |
Collapse
|
49
|
Brumberger ZL, Branch ME, Klein MW, Seals A, Shapiro MD, Vasu S. Cardiotoxicity risk factors with immune checkpoint inhibitors. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2022; 8:3. [PMID: 35277208 PMCID: PMC8915459 DOI: 10.1186/s40959-022-00130-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 02/21/2022] [Indexed: 12/18/2022]
Abstract
Background Checkpoint-inhibitor immunotherapies have had a profound effect in the treatment of cancer by inhibiting down-regulation of T-cell response to malignancy. The cardiotoxic potential of these agents was first described in murine models and, more recently, in numerous clinical case reports of pericarditis, myocarditis, pericardial effusion, cardiomyopathy, and new arrhythmias. The objective of our study was to determine the frequency of and associated risk factors for cardiotoxic events in patients treated with immune checkpoint inhibitors. Methods Medical records of patients who underwent immunotherapy with durvalumab, ipilimumab, nivolumab, and pembrolizumab at Wake Forest Baptist Health were reviewed. We collected retrospective data regarding sex, cancer type, age, and cardiovascular disease risk factors and medications. We aimed to identify new diagnoses of heart failure, atrial fibrillation, ventricular fibrillation/tachycardia, myocarditis, and pericarditis after therapy onset. To assess the relationship between CVD risk factors and the number of cardiac events, a multivariate model was applied using generalized linear regression. Incidence rate ratios were calculated for every covariate along with the adjusted P-value. We applied a multivariate model using logistic regression to assess the relationship between CVD risk factors and mortality. Odds ratios were calculated for every covariate along with the adjusted P-value. Adjusted P-values were calculated using multivariable regression adjusting for other covariates. Results Review of 538 medical records revealed the following events: 3 ventricular fibrillation/tachycardia, 12 pericarditis, 11 atrial fibrillation with rapid ventricular rate, 0 myocarditis, 8 heart failure. Significant risk factors included female gender, African American race, and tobacco use with IRR 3.34 (95% CI 1.421, 7.849; P = 0.006), IRR 3.39 (95% CI 1.141, 10.055; P = 0.028), and IRR 4.21 (95% CI 1.289, 13.763; P = 0.017) respectively. Conclusions Our study revealed 34 significant events, most frequent being pericarditis (2.2%) and atrial fibrillation (2.0%) with strongest risk factors being female gender, African American race, and tobacco use. Patients who meet this demographic, particularly those with planned pembrolizumab treatment, may benefit from early referral to a cardio-oncologist. Further investigation is warranted on the relationship between CTLA-4 and PD-L1 expression and cardiac adverse events with ICIs, particularly for these subpopulations.
Collapse
Affiliation(s)
- Zachary L Brumberger
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Mary E Branch
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Max W Klein
- Department of Internal Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA.
| | - Austin Seals
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Michael D Shapiro
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| | - Sujethra Vasu
- Department of Internal Medicine, Section On Cardiovascular Medicine, Wake Forest University School of Medicine, 1 Medical Center Boulevard, Winston-Salem, NC, 27157, USA
| |
Collapse
|
50
|
Hao Y, Cook MC. Inborn Errors of Immunity and Their Phenocopies: CTLA4 and PD-1. Front Immunol 2022; 12:806043. [PMID: 35154081 PMCID: PMC8832511 DOI: 10.3389/fimmu.2021.806043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022] Open
Abstract
Elucidating links between genotype and phenotype in patients with rare inborn errors of immunity (IEIs) provides insights into mechanisms of immune regulation. In many autosomal dominant IEIs, however, variation in expressivity and penetrance result in complex genotype-phenotype relations, while some autosomal recessive IEIs are so rare that it is difficult to draw firm conclusions. Phenocopies arise when an environmental or non-genetic factor replicates a phenotype conferred by a specific genotype. Phenocopies can result from therapeutic antibodies or autoantibodies that target a protein to replicate aspects of the phenotype conferred by mutations in the gene encoding the same protein. Here, we consider IEIs arising from rare genetic variants in CTLA4 and PDCD1 and compare clinical and laboratory manifestations arising as drug-induced phenocopies (immune related adverse events, IRAEs) in cancer patients treated with immune checkpoint inhibitors (ICI) and identify outstanding questions regarding mechanism of disease.
Collapse
Affiliation(s)
- Yuwei Hao
- Centre for Personalised Immunology and Department of Immunity and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia
| | - Matthew C Cook
- Centre for Personalised Immunology and Department of Immunity and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia.,Department of Immunology, Canberra Hospital, Woden, ACT, Australia
| |
Collapse
|