1
|
Sun L, Zhao Q, Ao S, Liu T, Wang Z, You J, Mi Z, Sun Y, Xue X, Ogese MO, Gardner J, Meng X, Naisbitt DJ, Liu H, Zhang F. Feedback regulation of VISTA and Treg by TNF-α controls T cell responses in drug allergy. Allergy 2024. [PMID: 39526799 DOI: 10.1111/all.16393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Severe cutaneous adverse reactions (SCARs) mediated by cytotoxic T lymphocytes are a series of life-threatening conditions with a mortality of 4%-20%. The clinical application of tumor necrosis factor-alpha (TNF-α) antagonist improves the outcome of some SCARs patients; however, this is complicated by the elusive and varied immunopathogenesis. AIM To investigate whether IgE antibody responses to HEMAs are associated with AD, its severity, and response to dupilumab. METHODS To clarify the precise process and optimize the therapy regimen of SCARs, we performed single-cell sequencing, in vitro functional and clinical analysis of patients with SCARs. RESULTS We observed that TNF-α breaks drug-specific T-cell tolerance by inhibiting the expression of V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA). Furthermore, TNF-α generated a positive feedback loop in the early phase of drug-specific T-cell activation, whereby B cells acted reciprocally on the corresponding T cells to reinforce TNF-α cytokine expression. In contrast, this pathway of TNF-α-VISTA signaling did not operate in memory effector T cells. Drug-specific memory effector T-cell responses were inhibited by increasing Treg cell expression in a negative feedback loop, with TNF-α antagonists preventing the inhibitory effect. These observations align with the clinical analysis that early but not late intervention with TNF-α antagonists significantly improved outcomes in SCARs patients. CONCLUSION Our findings defining feedback regulation of VISTA and Treg cells by TNF-α in different stages of the drug-specific T-cell response and, indicate that a Treg agonists, instead of TNF-α antagonists, could be used for treatment of patients with progressive SCARs.
Collapse
Affiliation(s)
- Lele Sun
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qing Zhao
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Suiting Ao
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tingting Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenzhen Wang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jiabao You
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zihao Mi
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yonghu Sun
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaotong Xue
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Monday O Ogese
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutic, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Joshua Gardner
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutic, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutic, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Dean J Naisbitt
- MRC Centre for Drug Safety Science, Department of Pharmacology and Therapeutic, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Hong Liu
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Furen Zhang
- Hospital for Skin Diseases, Shandong First Medical University, Jinan, Shandong, China
- Shandong Provincial Institute of Dermatology and Venereology, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
2
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [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: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
3
|
Choi SH, Chen YW, Panian J, Yuen K, McKay RR. Emerging innovative treatment strategies for advanced clear cell renal cell carcinoma. Oncologist 2024:oyae276. [PMID: 39401004 DOI: 10.1093/oncolo/oyae276] [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: 06/16/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Dramatic advances in biological discoveries, since the 1990s, have continued to reshape the treatment paradigm of metastatic renal cell carcinoma (RCC). Von Hippel Lindau (VHL) gene alterations are associated with pro-angiogenic activity and are central to the pathogenesis of clear cell RCC (ccRCC), the most predominant histologic subtype of RCC. Antiangiogenic strategies revolving around this VHL/HIF/VEGF axis have been shown to improve survival in metastatic ccRCC. The discovery of immune checkpoints and agents that target their inhibition introduced a new treatment paradigm for patients with RCC. While initially approved as monotherapy, studies investigating immune checkpoint inhibitor combinations have led to their approval as the new standard of care, providing durable responses and unprecedented improvements in clinical outcome. Despite these advances, the projected 14 390 deaths in 2024 from RCC underscore the need to continue efforts in expanding and optimizing treatment options for patients with metastatic RCC. This article reviews key findings that have transformed the way we understand and treat metastatic RCC, in addition to highlighting novel treatment strategies that are currently under development.
Collapse
Affiliation(s)
- Sharon H Choi
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Yu-Wei Chen
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Justine Panian
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
| | - Kit Yuen
- Department of Urology, University of California San Diego, San Diego, CA, United States
| | - Rana R McKay
- Division of Hematology Oncology, University of California San Diego, San Diego, CA, United States
- Department of Urology, University of California San Diego, San Diego, CA, United States
| |
Collapse
|
4
|
Azizi M, Mokhtari Z, Tavana S, Bemani P, Heidari Z, Ghazavi R, Rezaei M. A Comprehensive Study on the Prognostic Value and Clinicopathological Significance of Different Immune Checkpoints in Patients With Colorectal Cancer: A Systematic Review and Meta-Analysis. CURRENT THERAPEUTIC RESEARCH 2024; 101:100760. [PMID: 39434898 PMCID: PMC11492099 DOI: 10.1016/j.curtheres.2024.100760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 08/30/2024] [Indexed: 10/23/2024]
Abstract
Background The prognostic significance of immune checkpoint expression in the tumor microenvironment has been widely investigated in colorectal cancers. However, the results of these studies are inconsistent and limited to some immune checkpoints. Objective The study aimed to investigate the correlation between different immune checkpoint expression and clinicopathological features and prognostic parameters. Methods We conducted a systematic review and meta-analysis of the published literature in PubMed, Web of Science-Core Collection, Scopus, Embase, and Cochrane databases to summarize the association between various immune checkpoints expression on both tumor cells and immune cells with clinicopathological features and prognostic parameters in patients with colorectal cancer. Results One hundred four studies incorporating 22,939 patients were included in our meta-analysis. Our results showed that among the B7 family, the high expression of B7H3, B7H4, PD-1, and PD-L1 on tumor cells and tumor tissue was significantly associated with higher T stage, advanced tumor, node, metastasis (TNM) stage, presence of vascular invasion, and lymphatic invasion. In addition, patients with high expression of B7H3, B7H4, PD-1, PD-L1, and PD-L2 were associated with shorter overall survival. High expression of PD-1 and PD-L1 in immune cells correlated with the absence of lymph node metastasis, lower TNM stage, early T stage, poor overall survival, and disease-free survival, respectively. Moreover, we found significant positive correlations between CD70 and Galectin-3 expression with advanced T stage. HLA-II overexpression was correlated with the absence of lymph node metastasis (odds ratio = 0.21, 95% CI = 0.11-0.38, P < 0.001) and early TNM stage (odds ratio = 0.35, 95% CI = 0.26-0.47, P < 0.001). Conclusions Overexpression of B7H3, B7H4, PD-1, PD-L1, PD-L2, CD70, and Galectin-3 on tumors is significantly associated with unfavorable clinicopathological characteristics and poor prognostic factors. Hence, these immune checkpoints can serve as predictive biomarkers for prognosis and the clinicopathological features of colorectal cancer because this is essential to identify patients suitable for anticancer therapy with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Mahdieh Azizi
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Mokhtari
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shirin Tavana
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Bemani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Heidari
- Department of Biostatistics and Epidemiology, Faculty of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roghayeh Ghazavi
- Department of Knowledge and Information Science, Faculty of Education and Psychology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Marzieh Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
5
|
Kametani Y, Ito R, Manabe Y, Kulski JK, Seki T, Ishimoto H, Shiina T. PBMC-engrafted humanized mice models for evaluating immune-related and anticancer drug delivery systems. Front Mol Biosci 2024; 11:1447315. [PMID: 39228913 PMCID: PMC11368775 DOI: 10.3389/fmolb.2024.1447315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024] Open
Abstract
Immune-related drug delivery systems (DDSs) in humanized mouse models are at the forefront of cancer research and serve as bridges between preclinical studies and clinical applications. These systems offer unique platforms for exploring new therapies and understanding their interactions with human cells and the immune system. Here, we focus on a DDS and a peripheral blood mononuclear cell (PBMC)-engrafted humanized mouse model that we recently developed, and consider some of the key components, challenges, and applications to advance these systems towards better cancer treatment on the basis of a better understanding of the immune response. Our DDS is unique and has a dual function, an anticancer effect and a capacity to fine-tune the immune reaction. The PBL-NOG-hIL-4-Tg mouse system is superior to other available humanized mouse systems for the development of such multifunctional DDSs because it supports the rapid reconstruction of an individual donor's immunity and avoids the onset of graft-versus-host disease.
Collapse
Affiliation(s)
- Yoshie Kametani
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Japan
| | - Ryoji Ito
- Central Institute for Experimental Medicine and Life Science (CIEM), Kawasaki, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan
| | - Jerzy K. Kulski
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Faculty of Health and Medical Sciences, School of Biomedical Science, The University of Western Australia, Crawley, WA, Australia
| | - Toshiro Seki
- Department of Internal Medicine, Division of Nephrology, Endocrinology, and Metabolism, Tokai University School of Medicine, Isehara, Japan
| | - Hitoshi Ishimoto
- Department of Obstetrics and Gynecology, Tokai University School of Medicine, Isehara, Japan
| | - Takashi Shiina
- Department of Molecular Life Science, Division of Basic Medical Science, Tokai University School of Medicine, Isehara, Japan
- Institute of Advanced Biosciences, Tokai University, Hiratsuka, Japan
| |
Collapse
|
6
|
Ghaedrahmati F, Akbari V, Seyedhosseini-Ghaheh H, Esmaeil N. Strong capacity of differentiated PD-L1 CAR-modified UCB-CD34 + cells and PD-L1 CAR-modified UCB-CD34 +-derived NK cells in killing target cells and restoration of the anti-tumor function of PD-1-high exhausted T Cells. Stem Cell Res Ther 2024; 15:257. [PMID: 39135206 PMCID: PMC11321137 DOI: 10.1186/s13287-024-03871-5] [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: 05/13/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Using natural killer (NK) cells to treat hematopoietic and solid tumors has great promise. Despite their availability from peripheral blood and cord blood, stem cell-derived NK cells provide an "off-the-shelf" solution. METHODS In this study, we developed two CAR-NK cells targeting PD-L1 derived from lentiviral transduction of human umbilical cord blood (UCB)-CD34+ cells and UCB-CD34+-derived NK cells. The transduction efficiencies and in vitro cytotoxic functions including degranulation, cytokine production, and cancer cell necrosis of both resultants PD-L1 CAR-NK cells were tested in vitro on two different PD-L1 low and high-expressing solid tumor cell lines. RESULTS Differentiated CAR‑modified UCB-CD34+ cells exhibited enhanced transduction efficiency. The expression of anti-PD-L1 CAR significantly (P < 0.05) enhanced the cytotoxicity of differentiated CAR‑modified UCB-CD34+ cells and CAR-modified UCB-CD34+-derived NK cells against PD-L1 high-expressing tumor cell line. In addition, CAR-modified UCB-CD34+-derived NK cells significantly (P < 0.05) restored the tumor-killing ability of exhausted PD-1 high T cells. CONCLUSION Considering the more efficient transduction in stem cells and the possibility of producing CAR-NK cell products with higher yields, this approach is recommended for studies in the field of CAR-NK cells. Also, a pre-clinical study is now necessary to evaluate the safety and efficacy of these two CAR-NK cells individually and in combination with other therapeutic approaches.
Collapse
Affiliation(s)
- Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744, Iran.
- Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
- Pooya Zist-Mabna Hakim Company, Poursina Hakim Institute, Isfahan, Iran.
| |
Collapse
|
7
|
Joshi DC, Sharma A, Prasad S, Singh K, Kumar M, Sherawat K, Tuli HS, Gupta M. Novel therapeutic agents in clinical trials: emerging approaches in cancer therapy. Discov Oncol 2024; 15:342. [PMID: 39127974 PMCID: PMC11317456 DOI: 10.1007/s12672-024-01195-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host's immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, "precision medicine" customizes therapies to each patient's unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
Collapse
Affiliation(s)
- Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist., Ajmer, Rajasthan, India.
| | - Anurag Sharma
- Invertis Institute of Pharmacy, Invertis University Bareilly Uttar Pradesh, Bareilly, India
| | - Sonima Prasad
- Chandigarh University, Ludhiana-Chandigarh State Highway, Gharuan, Mohali, Punjab, 140413, India
| | - Karishma Singh
- Institute of Pharmaceutical Sciences, Faculty of Engineering and Technology, University of Lucknow, Lucknow, India
| | - Mayank Kumar
- Himalayan Institute of Pharmacy, Road, Near Suketi Fossil Park, Kala Amb, Hamidpur, Himachal Pradesh, India
| | - Kajal Sherawat
- Meerut Institute of Technology, Meerut, Uttar Pradesh, India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, India
| | - Madhu Gupta
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, India.
| |
Collapse
|
8
|
Jiang A, Li J, He Z, Liu Y, Qiao K, Fang Y, Qu L, Luo P, Lin A, Wang L. Renal cancer: signaling pathways and advances in targeted therapies. MedComm (Beijing) 2024; 5:e676. [PMID: 39092291 PMCID: PMC11292401 DOI: 10.1002/mco2.676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/09/2024] [Accepted: 07/15/2024] [Indexed: 08/04/2024] Open
Abstract
Renal cancer is a highlyheterogeneous malignancy characterized by rising global incidence and mortalityrates. The complex interplay and dysregulation of multiple signaling pathways,including von Hippel-Lindau (VHL)/hypoxia-inducible factor (HIF), phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), Hippo-yes-associated protein (YAP), Wnt/ß-catenin, cyclic adenosine monophosphate (cAMP), and hepatocyte growth factor (HGF)/c-Met, contribute to theinitiation and progression of renal cancer. Although surgical resection is thestandard treatment for localized renal cancer, recurrence and metastasiscontinue to pose significant challenges. Advanced renal cancer is associatedwith a poor prognosis, and current therapies, such as targeted agents andimmunotherapies, have limitations. This review presents a comprehensiveoverview of the molecular mechanisms underlying aberrant signaling pathways inrenal cancer, emphasizing their intricate crosstalk and synergisticinteractions. We discuss recent advancements in targeted therapies, includingtyrosine kinase inhibitors, and immunotherapies, such as checkpoint inhibitors.Moreover, we underscore the importance of multiomics approaches and networkanalysis in elucidating the complex regulatory networks governing renal cancerpathogenesis. By integrating cutting-edge research and clinical insights, this review contributesto the development of innovative diagnostic and therapeutic strategies, whichhave the potential to improve risk stratification, precision medicine, andultimately, patient outcomes in renal cancer.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Jinxin Li
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Ziwei He
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Ying Liu
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Kun Qiao
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Yu Fang
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Le Qu
- Department of UrologyJinling HospitalAffiliated Hospital of Medical SchoolNanjing UniversityNanjingChina
| | - Peng Luo
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Anqi Lin
- Department of OncologyZhujiang HospitalSouthern Medical UniversityGuangzhouGuangdongChina
| | - Linhui Wang
- Department of UrologyChanghai HospitalNaval Medical UniversityShanghaiChina
| |
Collapse
|
9
|
Vetsika EK, Fragoulis GE, Kyriakidi M, Verrou KM, Tektonidou MG, Alissafi T, Sfikakis PP. Insufficient PD-1 expression during active autoimmune responses: a deep single-cell proteomics analysis in inflammatory arthritis. Front Immunol 2024; 15:1403680. [PMID: 38911848 PMCID: PMC11190177 DOI: 10.3389/fimmu.2024.1403680] [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: 03/19/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
Objectives Programmed cell death protein-1 (PD-1) maintains peripheral immune tolerance by preventing T cell continuous activation. Aiming to understand the extent of PD-1 expression in inflammatory arthritis beyond its involvement with T cells, we assess its presence on various circulating single cells. Methods Mass cytometry analysis of patients with active seropositive/seronegative rheumatoid (RA; n=9/8) and psoriatic (PsA; n=9) arthritis versus healthy controls (HC; n=13), re-evaluating patients after 3 months of anti-rheumatic treatment. Results PD-1 was expressed in all leukocyte subpopulations, with the highest PD-1+ cell frequencies in eosinophils (59-73%) and T cells (50-60%), and the lowest in natural-killer cells (1-3%). PD-1+ cell frequencies and PD-1 median expression were comparable between patient subgroups and HC, in the majority of cell subsets. Exceptions included increases in certain T cell/B cell subsets of seropositive RA and specific monocyte subsets and dendritic cells of PsA; an expanded PD-1+CD4+CD45RA+CD27+CD28+ T subset, denoting exhausted T cells, was common across patient subgroups. Strikingly, significant inverse correlations between individual biomarkers of systemic inflammation (ESR and/or serum CRP) and PD-1+ cell frequencies and/or median expression were evident in several innate and adaptive immunity cell subsets of RA and PsA patients. Furthermore, all inverse correlations noted in individuals with active arthritis were no longer discernible in those who attained remission/low disease activity post-treatment. Conclusion PD-1 expression may be insufficient, relative to the magnitude of the concomitant systemic inflammatory response on distinct leukocyte subsets, varying between RA and PsA. Our results point to the potential therapeutic benefits of pharmacological PD-1 activation, to rebalance the autoimmune response and reduce inflammation.
Collapse
Affiliation(s)
- Eleni-Kyriaki Vetsika
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George E. Fragoulis
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Kyriakidi
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kleio-Maria Verrou
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria G. Tektonidou
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Themis Alissafi
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Laboratory of Immune Regulation, Center of Basic Sciences, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Petros P. Sfikakis
- Centre of New Biotechnologies and Precision Medicine (CNBPM), School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- First Department of Propaedeutic Internal Medicine and Joint Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Liu A, Zhang G, Yang Y, Xia Y, Li W, Liu Y, Cui Q, Wang D, Yu J. Two nomograms constructed for predicting the efficacy and prognosis of advanced non‑small cell lung cancer patients treated with anti‑PD‑1 inhibitors based on the absolute counts of lymphocyte subsets. Cancer Immunol Immunother 2024; 73:152. [PMID: 38833153 PMCID: PMC11150349 DOI: 10.1007/s00262-024-03738-x] [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: 03/10/2024] [Accepted: 05/17/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND Patients treated with immune checkpoint inhibitors (ICIs) are at risk of considerable adverse events, and the ongoing struggle is to accurately identify the subset of patients who will benefit. Lymphocyte subsets play a pivotal role in the antitumor response, this study attempted to combine the absolute counts of lymphocyte subsets (ACLS) with the clinicopathological parameters to construct nomograms to accurately predict the prognosis of advanced non-small cell lung cancer (aNSCLC) patients treated with anti-PD-1 inhibitors. METHODS This retrospective study included a training cohort (n = 200) and validation cohort (n = 100) with aNSCLC patients treated with anti-PD-1 inhibitors. Logistic and Cox regression were conducted to identify factors associated with efficacy and progression-free survival (PFS) respectively. Nomograms were built based on independent influencing factors, and assessed by the concordance index (C-index), calibration curve and receiver operating characteristic (ROC) curve. RESULT In training cohort, lower baseline absolute counts of CD3+ (P < 0.001) and CD4+ (P < 0.001) were associated with for poorer efficacy. Hepatic metastases (P = 0.019) and lower baseline absolute counts of CD3+ (P < 0.001), CD4+ (P < 0.001), CD8+ (P < 0.001), and B cells (P = 0.042) were associated with shorter PFS. Two nomograms to predict efficacy at 6-week after treatment and PFS at 4-, 8- and 12-months were constructed, and validated in validation cohort. The area under the ROC curve (AUC-ROC) of nomogram to predict response was 0.908 in training cohort and 0.984 in validation cohort. The C-index of nomogram to predict PFS was 0.825 in training cohort and 0.832 in validation cohort. AUC-ROC illustrated the nomograms had excellent discriminative ability. Calibration curves showed a superior consistence between the nomogram predicted probability and actual observation. CONCLUSION We constructed two nomogram based on ACLS to help clinicians screen of patients with possible benefit and make individualized treatment decisions by accurately predicting efficacy and PFS for advanced NSCLC patient treated with anti-PD-1 inhibitors.
Collapse
Affiliation(s)
- Aqing Liu
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guan Zhang
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjie Yang
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying Xia
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wentao Li
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunhe Liu
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qian Cui
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dong Wang
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianchun Yu
- Department of Oncology, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
11
|
Chen H, Zuo H, Huang J, Liu J, Jiang L, Jiang C, Zhang S, Hu Q, Lai H, Yin B, Yang G, Mai G, Li B, Chi H. Unravelling infiltrating T-cell heterogeneity in kidney renal clear cell carcinoma: Integrative single-cell and spatial transcriptomic profiling. J Cell Mol Med 2024; 28:e18403. [PMID: 39031800 PMCID: PMC11190954 DOI: 10.1111/jcmm.18403] [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: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 07/15/2024] Open
Abstract
Kidney renal clear cell carcinoma (KIRC) pathogenesis intricately involves immune system dynamics, particularly the role of T cells within the tumour microenvironment. Through a multifaceted approach encompassing single-cell RNA sequencing, spatial transcriptome analysis and bulk transcriptome profiling, we systematically explored the contribution of infiltrating T cells to KIRC heterogeneity. Employing high-density weighted gene co-expression network analysis (hdWGCNA), module scoring and machine learning, we identified a distinct signature of infiltrating T cell-associated genes (ITSGs). Spatial transcriptomic data were analysed using robust cell type decomposition (RCTD) to uncover spatial interactions. Further analyses included enrichment assessments, immune infiltration evaluations and drug susceptibility predictions. Experimental validation involved PCR experiments, CCK-8 assays, plate cloning assays, wound-healing assays and Transwell assays. Six subpopulations of infiltrating and proliferating T cells were identified in KIRC, with notable dynamics observed in mid- to late-stage disease progression. Spatial analysis revealed significant correlations between T cells and epithelial cells across varying distances within the tumour microenvironment. The ITSG-based prognostic model demonstrated robust predictive capabilities, implicating these genes in immune modulation and metabolic pathways and offering prognostic insights into drug sensitivity for 12 KIRC treatment agents. Experimental validation underscored the functional relevance of PPIB in KIRC cell proliferation, invasion and migration. Our study comprehensively characterizes infiltrating T-cell heterogeneity in KIRC using single-cell RNA sequencing and spatial transcriptome data. The stable prognostic model based on ITSGs unveils infiltrating T cells' prognostic potential, shedding light on the immune microenvironment and offering avenues for personalized treatment and immunotherapy.
Collapse
Affiliation(s)
- Haiqing Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haoyuan Zuo
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Jinbang Huang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Jie Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General SurgeryDazhou Central HospitalDazhouChina
| | - Lai Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Chenglu Jiang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Shengke Zhang
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Qingwen Hu
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Haotian Lai
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Bangchao Yin
- Department of PathologySixth People's Hospital of YibinYibinChina
| | - Guanhu Yang
- Department of Specialty MedicineOhio UniversityAthensOhioUSA
| | - Gang Mai
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
- Department of General Surgery (Hepatopancreatobiliary Surgery)Deyang People's HospitalDeyangChina
| | - Bo Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| | - Hao Chi
- School of Clinical Medicine, The Affiliated HospitalSouthwest Medical UniversityLuzhouChina
| |
Collapse
|
12
|
Kim D, Javius-Jones K, Mamidi N, Hong S. Dendritic nanoparticles for immune modulation: a potential next-generation nanocarrier for cancer immunotherapy. NANOSCALE 2024; 16:10208-10220. [PMID: 38727407 DOI: 10.1039/d4nr00635f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Immune activation, whether occurring from direct immune checkpoint blockade or indirectly as a result of chemotherapy, is an approach that has drastically impacted the way we treat cancer. Utilizing patients' own immune systems for anti-tumor efficacy has been translated to robust immunotherapies; however, clinically significant successes have been achieved in only a subset of patient populations. Dendrimers and dendritic polymers have recently emerged as a potential nanocarrier platform that significantly improves the therapeutic efficacy of current and next-generation cancer immunotherapies. In this paper, we highlight the recent progress in developing dendritic polymer-based therapeutics with immune-modulating properties. Specifically, dendrimers, dendrimer hybrids, and dendronized copolymers have demonstrated promising results and are currently in pre-clinical development. Despite their early stage of development, these nanocarriers hold immense potential to make profound impact on cancer immunotherapy and combination therapy. This overview provides insights into the potential impact of dendrimers and dendron-based polymers, offering a preview of their potential utilities for various aspects of cancer treatment.
Collapse
Affiliation(s)
- DaWon Kim
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Kaila Javius-Jones
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
| | - Narsimha Mamidi
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, School of Pharmacy, Madison, WI, USA.
- Wisconsin Center for NanoBioSystems, University of Wisconsin-Madison, Madison, WI, USA
- Lachman Institute for Drug Development, University of Wisconsin-Madison, Madison, WI, USA
- Yonsei Frontier Lab, Yonsei University, Seoul, Korea
| |
Collapse
|
13
|
Kim SM, Park N, Park HB, Lee J, Chun C, Kim KH, Choi JS, Kim HJ, Choi S, Lee JH. Exploring novel immunotherapy biomarker candidates induced by cancer deformation. PLoS One 2024; 19:e0303433. [PMID: 38743676 PMCID: PMC11093347 DOI: 10.1371/journal.pone.0303433] [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: 11/10/2023] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) demands urgent attention for the development of effective treatment strategies due to its aggressiveness and limited therapeutic options [1]. This research is primarily focused on identifying new biomarkers vital for immunotherapy, with the aim of developing tailored treatments specifically for TNBC, such as those targeting the PD-1/PD-L1 pathway. To achieve this, the study places a strong emphasis on investigating Ig genes, a characteristic of immune checkpoint inhibitors, particularly genes expressing Ig-like domains with altered expression levels induced by "cancer deformation," a condition associated with cancer malignancy. Human cells can express approximately 800 Ig family genes, yet only a few Ig genes, including PD-1 and PD-L1, have been developed into immunotherapy drugs thus far. Therefore, we investigated the Ig genes that were either upregulated or downregulated by the artificial metastatic environment in TNBC cell line. As a result, we confirmed the upregulation of approximately 13 Ig genes and validated them using qPCR. In summary, our study proposes an approach for identifying new biomarkers applicable to future immunotherapies aimed at addressing challenging cases of TNBC where conventional treatments fall short.
Collapse
Affiliation(s)
- Se Min Kim
- Life Science and Biotechnology Department (LSBT), Underwood Division (UD), Underwood International College, Yonsei University, Sinchon, Seoul, Korea
| | - Namu Park
- Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, Washington, United States of America
| | - Hye Bin Park
- Digital Health Care Research Center, Gumi Electronics and Information Technology Research Institute (GERI), Gumidaero, Gumi, Gyeongbuk, South Korea
| | - JuKyung Lee
- Digital Health Care Research Center, Gumi Electronics and Information Technology Research Institute (GERI), Gumidaero, Gumi, Gyeongbuk, South Korea
| | - Changho Chun
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington, United States of America
| | - Kyung Hoon Kim
- Department of Bioengineering, University of Washington, Seattle, Washington, United States of America
| | - Jong Seob Choi
- Division of Advanced Materials Engineering, Kongju National University, Chungnam, South Korea
| | - Hyung Jin Kim
- School of Electrical & Electronic Engineerin, Ulsan College, Ulsan, Korea
| | - Sekyu Choi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Jung Hyun Lee
- Department of Dermatology, School of Medicine, University of Washington, Seattle, Washington, United States of America
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
14
|
Patwekar M, Sehar N, Patwekar F, Medikeri A, Ali S, Aldossri RM, Rehman MU. Novel immune checkpoint targets: A promising therapy for cancer treatments. Int Immunopharmacol 2024; 126:111186. [PMID: 37979454 DOI: 10.1016/j.intimp.2023.111186] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
The immune system frequently comprises immunological checkpoints. They serve as a barrier to keep the immune system from overreacting and damaging cells that are robust. Immune checkpoint inhibitors (ICIs) are utilized in immunotherapy to prevent the synergy of partner proteins of checkpoint proteins with auxiliary proteins. Moreover, the T cells may target malignant cells since the "off" signal cannot be conveyed. ICIs, which are mostly composed of monoclonal antibodies (mAbs) against cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and anti- programmed death-1/programmed ligand 1 (anti-PD-1/PD-L1), might transform the context of cancer therapy. Further, more patients continued to exhibit adaptive resistance, even though several ICIs demonstrated convincing therapeutic benefits in selective tumor types. Immune checkpoint therapy's overall effectiveness is still lacking at this time. A popular area of study involves investigating additional immune checkpoint molecules. Recent research has found a number of fresh immune checkpoint targets, including NKG2A ligands, TIGIT, B7-H6 ligands, Galectin 3, TIM3, and so on. These targets have been focus of the study, and recent investigational approaches have shown encouraging outcomes. In this review article, we covered the development and present level understanding of these recently identified immune checkpoint molecules, its effectiveness and limitations.
Collapse
Affiliation(s)
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, 110062, India
| | - Faheem Patwekar
- Luqman College of Pharmacy, Gulbarga, 585102, Karnataka, India
| | | | - Shafat Ali
- Cytogenetics and Molecular Biology Laboratory, Centre of Research for Development, University of Kashmir, Srinagar, 190006, Jammu and Kashmir, India.
| | - Rana M Aldossri
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
15
|
Tsai SC, Farn SS, Lo WL, Ou Yang FY, Kang YC, Chen LC, Chen KT, Liao JW, Kung JY, Chen JT, Huang FYJ. Evaluation of Chelator-to-Antibody Ratio on Development of 89Zr-iPET Tracer for Imaging of PD-L1 Expression on Tumor. Int J Mol Sci 2023; 24:17132. [PMID: 38138961 PMCID: PMC10743313 DOI: 10.3390/ijms242417132] [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: 10/25/2023] [Revised: 11/23/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023] Open
Abstract
89Zr-iPET has been widely used for preclinical and clinical immunotherapy studies to predict patient stratification or evaluate therapeutic efficacy. In this study, we prepared and evaluated 89Zr-DFO-anti-PD-L1-mAb tracers with varying chelator-to-antibody ratios (CARs), including 89Zr-DFO-anti-PD-L1-mAb_3X (tracer_3X), 89Zr-DFO-anti-PD-L1-mAb_10X (tracer_10X), and 89Zr-DFO-anti-PD-L1-mAb_20X (tracer_20X). The DFO-anti-PD-L1-mAb conjugates with varying CARs were prepared using a random conjugation method and then subjected to quality control. The conjugates were radiolabeled with 89Zr and evaluated in a PD-L1-expressing CT26 tumor-bearing mouse model. Next, iPET imaging, biodistribution, pharmacokinetics, and ex vivo pathological and immunohistochemical examinations were conducted. LC-MS analysis revealed that DFO-anti-PD-L1-mAb conjugates were prepared with CARs ranging from 0.4 to 2.0. Radiochemical purity for all tracer groups was >99% after purification. The specific activity levels of tracer_3X, tracer_10X, and tracer_20X were 2.2 ± 0.6, 8.2 ± 0.6, and 10.5 ± 1.6 μCi/μg, respectively. 89Zr-iPET imaging showed evident tumor uptake in all tracer groups and reached the maximum uptake value at 24 h postinjection (p.i.). Biodistribution data at 168 h p.i. revealed that the tumor-to-liver, tumor-to-muscle, and tumor-to-blood uptake ratios for tracer_3X, tracer_10X, and tracer_20X were 0.46 ± 0.14, 0.58 ± 0.33, and 1.54 ± 0.51; 4.7 ± 1.3, 7.1 ± 3.9, and 14.7 ± 1.1; and 13.1 ± 5.8, 19.4 ± 13.8, and 41.3 ± 10.6, respectively. Significant differences were observed between tracer_3X and tracer_20X in the aforementioned uptake ratios at 168 h p.i. The mean residence time and elimination half-life for tracer_3X, tracer_10X, and tracer_20X were 25.4 ± 4.9, 24.2 ± 6.1, and 25.8 ± 3.3 h and 11.8 ± 0.5, 11.1 ± 0.7, and 11.7 ± 0.6 h, respectively. No statistical differences were found between-tracer in the aforementioned pharmacokinetic parameters. In conclusion, 89Zr-DFO-anti-PD-L1-mAb tracers with a CAR of 1.4-2.0 may be better at imaging PD-L1 expression in tumors than are traditional low-CAR 89Zr-iPET tracers.
Collapse
Affiliation(s)
- Shih-Chuan Tsai
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan; (S.-C.T.); (J.-Y.K.)
| | - Shiou-Shiow Farn
- National Atomic Research Institute, Taoyuan 325207, Taiwan; (S.-S.F.); (W.-L.L.); (F.-Y.O.Y.); (L.-C.C.); (J.-T.C.)
| | - Wei-Lin Lo
- National Atomic Research Institute, Taoyuan 325207, Taiwan; (S.-S.F.); (W.-L.L.); (F.-Y.O.Y.); (L.-C.C.); (J.-T.C.)
| | - Fang-Yu Ou Yang
- National Atomic Research Institute, Taoyuan 325207, Taiwan; (S.-S.F.); (W.-L.L.); (F.-Y.O.Y.); (L.-C.C.); (J.-T.C.)
| | - Yong-Ching Kang
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan;
| | - Liang-Cheng Chen
- National Atomic Research Institute, Taoyuan 325207, Taiwan; (S.-S.F.); (W.-L.L.); (F.-Y.O.Y.); (L.-C.C.); (J.-T.C.)
| | - Kuo-Ting Chen
- Department of Chemistry, National Dong Hwa University, Hualien 974301, Taiwan;
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, National Chung-Hsing University, Taichung 402202, Taiwan;
| | - Jui-Yin Kung
- Department of Nuclear Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan; (S.-C.T.); (J.-Y.K.)
| | - Jenn-Tzong Chen
- National Atomic Research Institute, Taoyuan 325207, Taiwan; (S.-S.F.); (W.-L.L.); (F.-Y.O.Y.); (L.-C.C.); (J.-T.C.)
| | - Feng-Yun J. Huang
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung 406053, Taiwan;
| |
Collapse
|
16
|
Calvo-Barreiro L, Talagayev V, Pach S, Abdel-Rahman SA, Wolber G, Gabr MT. Discovery of ICOS-Targeted Small Molecules Using Pharmacophore-Based Screening. ChemMedChem 2023; 18:e202300305. [PMID: 37845178 DOI: 10.1002/cmdc.202300305] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
There are currently no small molecules clinically approved as immune checkpoint modulators. Besides possessing oral bioavailability, cell-penetrating capabilities and enhanced tumor penetration compared to monoclonal antibodies (mAbs), small molecules are amenable to pharmacokinetic optimization, which allows adopting flexible dosage regimens that may avoid immune-related adverse events associated with mAbs. The interaction of inducible co-stimulator (ICOS) with its ligand (ICOS-L) plays key roles in T-cell differentiation and activation of T-cell to B-cell functions. This study represents the development and validation of a virtual screening strategy to identify small molecules that bind a novel druggable binding pocket in human ICOS. We used a lipophilic canyon in the apo-structure of ICOS and the ICOS/ICOS-L interface individually as templates for molecular dynamics simulation to generate 3D pharmacophores subsequently used for virtual screening campaigns. Our strategy was successful finding a first-in-class small molecule ICOS binder (5P, KD value=108.08±26.76 μM) and validating biophysical screening platforms for ICOS-targeted small molecules. We anticipate that future structural optimization of 5P will result in the discovery of high affinity chemical ligands for ICOS.
Collapse
Affiliation(s)
- Laura Calvo-Barreiro
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY-10065, USA
| | - Valerij Talagayev
- Molecular Design Lab, Department of Chemistry, Biology and Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Königin-Luisestr. 2+4, 14195, Berlin, Germany
| | - Szymon Pach
- Molecular Design Lab, Department of Chemistry, Biology and Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Königin-Luisestr. 2+4, 14195, Berlin, Germany
| | - Somaya A Abdel-Rahman
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY-10065, USA
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Gerhard Wolber
- Molecular Design Lab, Department of Chemistry, Biology and Pharmacy, Institute of Pharmacy, Freie Universität Berlin, Königin-Luisestr. 2+4, 14195, Berlin, Germany
| | - Moustafa T Gabr
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, NY-10065, USA
| |
Collapse
|
17
|
Cui Q, Li W, Wang D, Wang S, Yu J. Prognostic significance of blood-based PD-L1 analysis in patients with non-small cell lung cancer undergoing immune checkpoint inhibitor therapy: a systematic review and meta-analysis. World J Surg Oncol 2023; 21:318. [PMID: 37821941 PMCID: PMC10566159 DOI: 10.1186/s12957-023-03215-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/03/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND The main types of PD-L1 in the blood include soluble PD-L1 (sPD-L1), exosomal PD-L1 (exoPD-L1), and PD-L1 in circulating tumor cells (CTCs). However, the predictive and prognostic values of these three indicators in patients with non-small cell lung cancer (NSCLC) undergoing immune checkpoint inhibitor (ICI) therapy are unclear, warranting a systematic meta-analysis. METHODS A systematic literature search was performed in the PubMed, Cochrane Library, and Embase databases. The pooled hazard ratio (HR) and 95% confidence interval (CI) values were extracted from the included studies to investigate the correlation between the three PD-L1 indicators and overall survival (OS) or progression-free survival (PFS). The Newcastle-Ottawa Scale (NOS) was used to examine the quality of the included studies. Subgroup analyses were employed to investigate the heterogeneity. The publication bias of the included studies was assessed using Begg's and Egger's tests. P < 0.05 was regarded as significantly different. RESULTS The pooled results revealed that high pre-treatment sPD-L1 levels were significantly associated with inferior OS (HR = 2.32, 95% CI = 1.68-3.18, P < 0.001) and PFS (HR = 2.52, 95% CI = 1.72-3.68, P < 0.001). However, dynamic changes in sPD-L1 after immunotherapy were not statistically significant for OS (HR = 1.46, 95% CI = 0.65-3.26, P > 0.05) or PFS (HR = 1.62, 95% CI = 0.92-2.86, P > 0.05). Meanwhile, the upregulated pre-treatment exoPD-L1 levels were significantly associated with poor PFS (HR = 4.44, 95% CI = 2.87-6.89, P < 0.001), whereas the post-treatment dynamic upregulation of exoPD-L1 was significantly correlated with superior PFS (HR = 0.36, 95% CI = 0.24-0.54, P < 0.001) and OS (HR = 0.20, 95% CI = 0.07-0.53, P < 0.001). For PD-L1 in CTCs, the pooled results indicated that PD-L1 expression in CTCs was not significantly correlated with OS (HR = 0.75, 95% CI = 0.49-1.13, P = 0.170) and PFS (HR = 0.79, 95% CI = 0.59-1.06, P = 0.12). CONCLUSIONS Blood-based PD-L1 analysis is a potential strategy for predicting treatment efficacy and prognosis in patients with cancer.
Collapse
Affiliation(s)
- Qian Cui
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wentao Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dong Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shuangcui Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jianchun Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
18
|
Sharma D, Xuan Leong K, Palhares D, Czarnota GJ. Radiation combined with ultrasound and microbubbles: A potential novel strategy for cancer treatment. Z Med Phys 2023; 33:407-426. [PMID: 37586962 PMCID: PMC10517408 DOI: 10.1016/j.zemedi.2023.04.007] [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: 01/11/2023] [Revised: 03/31/2023] [Accepted: 04/11/2023] [Indexed: 08/18/2023]
Abstract
Cancer is one of the leading causes of death worldwide. Several emerging technologies are helping to battle cancer. Cancer therapies have been effective at killing cancer cells, but a large portion of patients still die to this disease every year. As such, more aggressive treatments of primary cancers are employed and have been shown to be capable of saving a greater number of lives. Recent research advances the field of cancer therapy by employing the use of physical methods to alter tumor biology. It uses microbubbles to enhance radiation effect by damaging tumor vasculature followed by tumor cell death. The technique can specifically target tumor volumes by conforming ultrasound fields capable of microbubbles stimulation and localizing it to avoid vascular damage in surrounding tissues. Thus, this new application of ultrasound-stimulated microbubbles (USMB) can be utilized as a novel approach to cancer therapy by inducing vascular disruption resulting in tumor cell death. Using USMB alongside radiation has showed to augment the anti-vascular effect of radiation, resulting in enhanced tumor response. Recent work with nanobubbles has shown vascular permeation into intracellular space, extending the use of this new treatment method to potentially further improve the therapeutic effect of the ultrasound-based therapy. The significant enhancement of localized tumor cell kill means that radiation-based treatments can be made more potent with lower doses of radiation. This technique can manifest a greater impact on radiation oncology practice by increasing treatment effectiveness significantly while reducing normal tissue toxicity. This review article summarizes the past and recent advances in USMB enhancement of radiation treatments. The review mainly focuses on preclinical findings but also highlights some clinical findings that use USMB as a therapeutic modality in cancer therapy.
Collapse
Affiliation(s)
- Deepa Sharma
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Kai Xuan Leong
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Daniel Palhares
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Gregory J Czarnota
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada; Departments of Radiation Oncology, and Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
Śledź M, Wojciechowska A, Zagożdżon R, Kaleta B. In Situ Programming of CAR-T Cells: A Pressing Need in Modern Immunotherapy. Arch Immunol Ther Exp (Warsz) 2023; 71:18. [PMID: 37419996 PMCID: PMC10329070 DOI: 10.1007/s00005-023-00683-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/05/2023] [Indexed: 07/09/2023]
Abstract
Chimeric antigen receptor-T (CAR-T) cell-based therapy has become a successful option for treatment of numerous hematological malignancies, but also raises hope in a range of non-malignant diseases. However, in a traditional approach, generation of CAR-T cells is associated with the separation of patient's lymphocytes, their in vitro modification, and expansion and infusion back into patient's bloodstream. This classical protocol is complex, time-consuming, and expensive. Those problems could be solved by successful protocols to produce CAR-T cells, but also CAR-natural killer cells or CAR macrophages, in situ, using viral platforms or non-viral delivery systems. Moreover, it was demonstrated that in situ CAR-T induction may be associated with reduced risk of the most common toxicities associated with CAR-T therapy, such as cytokine release syndrome, immune effector cell-associated neurotoxicity syndrome, and "on-target, off-tumor" toxicity. This review aims to summarize the current state-of-the-art and future perspectives for the in situ-produced CAR-T cells. Indeed, preclinical work in this area, including animal studies, raises hope for prospective translational development and validation in practical medicine of strategies for in situ generation of CAR-bearing immune effector cells.
Collapse
Affiliation(s)
- Marta Śledź
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | - Radosław Zagożdżon
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Beata Kaleta
- Department of Clinical Immunology, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
20
|
Alturki NA. Review of the Immune Checkpoint Inhibitors in the Context of Cancer Treatment. J Clin Med 2023; 12:4301. [PMID: 37445336 DOI: 10.3390/jcm12134301] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Checkpoint proteins are an integral part of the immune system and are used by the tumor cells to evade immune response, which helps them grow uncontrollably. By blocking these proteins, immune checkpoint inhibitors can restore the capability of the immune system to attack cancer cells and stop their growth. These findings are backed by adequate clinical trial data and presently, several FDA-approved immune checkpoint inhibitors exist in the market for treating various types of cancers, including melanoma, hepatocellular, endometrial, lung, kidney and others. Their mode of action is inhibition by targeting the checkpoint proteins CTLA-4, PD-1, PD-L1, etc. They can be used alone as well as in amalgamation with other cancer treatments, like surgery, radiation or chemotherapy. Since these drugs target only specific immune system proteins, their side effects are reduced in comparison with the traditional chemotherapy drugs, but may still cause a few affects like fatigue, skin rashes, and fever. In rare cases, these inhibitors are known to have caused more serious side effects, such as cardiotoxicity, and inflammation in the intestines or lungs. Herein, we provide an overview of these inhibitors and their role as biomarkers, immune-related adverse outcomes and clinical studies in the treatment of various cancers, as well as present some future perspectives.
Collapse
Affiliation(s)
- Norah A Alturki
- Clinical Laboratory Science Department, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
21
|
Swebocki T, Barras A, Abderrahmani A, Haddadi K, Boukherroub R. Deep Eutectic Solvents Comprising Organic Acids and Their Application in (Bio)Medicine. Int J Mol Sci 2023; 24:ijms24108492. [PMID: 37239842 DOI: 10.3390/ijms24108492] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Over the last years, we observed a significant increase in the number of published studies that focus on the synthesis and characterization of deep eutectic solvents (DESs). These materials are of particular interest mainly due to their physical and chemical stability, low vapor pressure, ease of synthesis, and the possibility of tailoring their properties through dilution or change of the ratio of parent substances (PS). DESs, considered as one of the greenest families of solvents, are used in many fields, such as organic synthesis, (bio)catalysis, electrochemistry, and (bio)medicine. DESs applications have already been reported in various review articles. However, these reports mainly described these components' basics and general properties without focusing on the particular, PS-wise, group of DESs. Many DESs investigated for potential (bio)medical applications comprise organic acids. However, due to the different aims of the reported studies, many of these substances have not yet been investigated thoroughly, which makes it challenging for the field to move forward. Herein, we propose distinguishing DESs comprising organic acids (OA-DESs) as a specific group derived from natural deep eutectic solvents (NADESs). This review aims to highlight and compare the applications of OA-DESs as antimicrobial agents and drug delivery enhancers-two essential fields in (bio)medical studies where DESs have already been implemented and proven their potential. From the survey of the literature data, it is evident that OA-DESs represent an excellent type of DESs for specific biomedical applications, owing to their negligible cytotoxicity, fulfilling the rules of green chemistry and being generally effective as drug delivery enhancers and antimicrobial agents. The main focus is on the most intriguing examples and (where possible) application-based comparison of particular groups of OA-DESs. This should highlight the importance of OA-DESs and give valuable clues on the direction the field can take.
Collapse
Affiliation(s)
- Tomasz Swebocki
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN-Institut d'Electronique de Microélectronique et de Nanotechnologie, 59000 Lille, France
| | - Alexandre Barras
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN-Institut d'Electronique de Microélectronique et de Nanotechnologie, 59000 Lille, France
| | - Amar Abderrahmani
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN-Institut d'Electronique de Microélectronique et de Nanotechnologie, 59000 Lille, France
| | - Kamel Haddadi
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN-Institut d'Electronique de Microélectronique et de Nanotechnologie, 59000 Lille, France
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Centrale Lille, Univ. Polytechnique Hauts-de-France, UMR 8520-IEMN-Institut d'Electronique de Microélectronique et de Nanotechnologie, 59000 Lille, France
| |
Collapse
|
22
|
Gupta R, Kadhim MM, Turki Jalil A, Qasim Alasheqi M, Alsaikhan F, Khalimovna Mukhamedova N, Alexis Ramírez-Coronel A, Hassan Jawhar Z, Ramaiah P, Najafi M. The interactions of docetaxel with tumor microenvironment. Int Immunopharmacol 2023; 119:110214. [PMID: 37126985 DOI: 10.1016/j.intimp.2023.110214] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/16/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
There are several interactions within the tumor microenvironment (TME) that affect the response of cancer cells to therapy. There are also a large number of cells and secretions in TME that increase resistance to therapy. Following the release of immunosuppressive, pro-angiogenic, and metastatic molecules by certain cells such as tumor-associated macrophages (TAMs), cancer-associated fibroblasts (CAFs), and cancer cells, immune evasion, angiogenesis, and metastasis may be induced. However, natural killer (NK) cells and cytotoxic CD8 + T lymphocytes (CTLs) can responsively release anticancer molecules. In addition, anticancer drugs can modulate these cells and their interactions in favor of either cancer resistance or therapy. Docetaxel belongs to taxanes, a class of anti-tumor drugs, which acts through the polymerization of tubulin and the induction of cell cycle arrest. Also, it has been revealed that taxanes including docetaxel affect cancer cells and the other cells within TME through some other mechanisms such as modulation of immune system responses, angiogenesis, and metastasis. In this paper, we explain the basic mechanisms of docetaxel interactions with malignant cells. Besides, we review the diverse effects of docetaxel on TME and cancer cells in consequence. Lastly, the modulatory effects of docetaxel alone or in conjunction with other anticancer agents on anti-tumor immunity, cancer cell resistance, angiogenesis, and metastasis will be discussed.
Collapse
Affiliation(s)
- Reena Gupta
- Institute of Pharmaceutical Research, GLA University, District-Mathura, 281406 U. P., India
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad 10022, Iraq
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla 51001, Iraq.
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia.
| | | | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Cuenca 010107, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Medillin 050001, Colombia; Educational Statistics Research Group (GIEE), National University of Education, Azogues 030102, Ecuador
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil 44001, Iraq; Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil 44001, Iraq
| | | | - Masoud Najafi
- Medical Technology Research Center, Institute of Health Technology, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran.
| |
Collapse
|
23
|
Elalouf A. Infections after organ transplantation and immune response. Transpl Immunol 2023; 77:101798. [PMID: 36731780 DOI: 10.1016/j.trim.2023.101798] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 01/08/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023]
Abstract
Organ transplantation has provided another chance of survival for end-stage organ failure patients. Yet, transplant rejection is still a main challenging factor. Immunosuppressive drugs have been used to avoid rejection and suppress the immune response against allografts. Thus, immunosuppressants increase the risk of infection in immunocompromised organ transplant recipients. The infection risk reflects the relationship between the nature and severity of immunosuppression and infectious diseases. Furthermore, immunosuppressants show an immunological impact on the genetics of innate and adaptive immune responses. This effect usually reactivates the post-transplant infection in the donor and recipient tissues since T-cell activation has a substantial role in allograft rejection. Meanwhile, different infections have been found to activate the T-cells into CD4+ helper T-cell subset and CD8+ cytotoxic T-lymphocyte that affect the infection and the allograft. Therefore, the best management and preventive strategies of immunosuppression, antimicrobial prophylaxis, and intensive medical care are required for successful organ transplantation. This review addresses the activation of immune responses against different infections in immunocompromised individuals after organ transplantation.
Collapse
Affiliation(s)
- Amir Elalouf
- Bar-Ilan University, Department of Management, Ramat Gan 5290002, Israel.
| |
Collapse
|
24
|
Sánchez-León ML, Jiménez-Cortegana C, Silva Romeiro S, Garnacho C, de la Cruz-Merino L, García-Domínguez DJ, Hontecillas-Prieto L, Sánchez-Margalet V. Defining the Emergence of New Immunotherapy Approaches in Breast Cancer: Role of Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:5208. [PMID: 36982282 PMCID: PMC10048951 DOI: 10.3390/ijms24065208] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Breast cancer (BC) continues to be the most diagnosed tumor in women and a very heterogeneous disease both inter- and intratumoral, mainly given by the variety of molecular profiles with different biological and clinical characteristics. Despite the advancements in early detection and therapeutic strategies, the survival rate is low in patients who develop metastatic disease. Therefore, it is mandatory to explore new approaches to achieve better responses. In this regard, immunotherapy arose as a promising alternative to conventional treatments due to its ability to modulate the immune system, which may play a dual role in this disease since the relationship between the immune system and BC cells depends on several factors: the tumor histology and size, as well as the involvement of lymph nodes, immune cells, and molecules that are part of the tumor microenvironment. Particularly, myeloid-derived suppressor cell (MDSC) expansion is one of the major immunosuppressive mechanisms used by breast tumors since it has been associated with worse clinical stage, metastatic burden, and poor efficacy of immunotherapies. This review focuses on the new immunotherapies in BC in the last five years. Additionally, the role of MDSC as a therapeutic target in breast cancer will be described.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carlos Jiménez-Cortegana
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Silvia Silva Romeiro
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Víctor Sánchez-Margalet
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| |
Collapse
|
25
|
Rashti A, Akbari V. Construction and Periplasmic Expression of a Bispecific Tandem scFv for Dual Targeting of Immune Checkpoints. Adv Biomed Res 2023; 12:42. [PMID: 37057231 PMCID: PMC10086663 DOI: 10.4103/abr.abr_31_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 05/21/2022] [Accepted: 05/22/2022] [Indexed: 04/15/2023] Open
Abstract
Background Immune checkpoints are molecules that act as regulators of immune system pathways. However, some tumor cells can express the ligands of immune checkpoints to escape from antitumor immune responses. Some agents, such as antibodies, can inhibit these checkpoints that prevent the immune system from targeting and killing cancer cells. The aim of this study was to express a novel bispecific tandem scFv in periplasmic space of Escherichia coli for simultaneous targeting of two immune checkpoints, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). Materials and Methods The bispecific tandem scFv was constructed based on the variable regions gene of anti-PD1 and anti-CTLA-4 antibodies. The optimum codon for expression in E. coli was chemically synthesized and subcloned in periplasmic expression plasmid. After transformation, the effect of cultivation conditions on periplasmic expression of the protein in E. coli BL21(DE3) was evaluated. Then, the bispecific tandem scFv was purified and its binding ability to cells expressing PD-1 and CTLA-4 was evaluated. Results Expression of tandem scFv with a molecular weight of 55 kDa was verified by Sodium dodecyl sulfate-polyacrylamide gel electrophoresis and western blotting analysis. The best condition for soluble periplasmic expression was obtained to be incubation with 0.5 mM isopropyl β-D-1-thiogalactopyranoside at 23°C. The protein was successfully purified using affinity chromatography with a final yield of 4.5 mg/L. Binding analysis confirmed the bioactivity of purified the tandem scFv. Conclusion This bispecific tandem scFv could be a potential candidate to cancer immunotherapy, although more biological activity assessments are still required to be carried out.
Collapse
Affiliation(s)
- Amirreza Rashti
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
- Address for correspondence: Dr. Vajihe Akbari, Department of Pharmaceutical Biotechnology, Isfahan University of Medical Sciences, Isfahan, Iran. E-mail:
| |
Collapse
|
26
|
Liu A, Zhang G, Yang Y, Xia Y, Li W, Liu Y, Cui Q, Wang D, Zhao J, Yu J. A clinical nomogram based on absolute count of lymphocyte subsets for predicting overall survival in patients with non-small cell lung cancer. Int Immunopharmacol 2023; 114:109391. [PMID: 36508919 DOI: 10.1016/j.intimp.2022.109391] [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: 06/13/2022] [Revised: 08/26/2022] [Accepted: 10/24/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The absolute count of lymphocyte subsets (ACLS) is correlated to the prognosis of multiple malignancies. This study aimed to combine the ACLS with the clinicopathological parameters to develop a nomogram to accurately predict the prognosis of non-small cell lung cancer (NSCLC) patients. METHODS This retrospective study included a training cohort (n = 1685) and validation cohort (n = 337) with NSCLC patients treated in First Teaching Hospital of Tianjin University of Traditional Chinese Medicine between January 2018 and January 2021. Cox regression were conducted to identify factors associated with overall survival. The nomogram was built based on 10 significant factors, and evaluated by the concordance index (C-index), calibration curve and receiver operating characteristic (ROC) curve. RESULTS In the training cohort, the multivariate cox proportional hazard regression analysis showed that the independent factors for overall survival (OS) included age, brain metastases, hepatic metastases, respiratory system diseases, clinical stages, surgery, absolute count (AC) of CD3+, CD4+, CD8+, and NK cells, which were all applied in the nomogram. The C-index of the nomogram to predict OS was 0.777 (95% CI, 0.751-0.802) in training cohort and 0.822 (95% CI, 0.798-0.846) in validation cohort. The area under the ROC showed a good discriminative ability in both cohorts. Calibration curves presented an excellent consistence between the nomogram predicted probability and actual observation. CONCLUSIONS We established a prognostic nomogram to predict OS of the NSCLC patient. This nomogram provided a more quantitative, scientific and objective basis for accurate diagnosis and individual management of NSCLC patients.
Collapse
Affiliation(s)
- Aqing Liu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guan Zhang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanjie Yang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying Xia
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wentao Li
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yunhe Liu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Qian Cui
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Dong Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China; Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jian Zhao
- Department of Oncology-Pathology, Karolinska Institutet, BioClinicum, Karolinska University Hospital Solna, Stockholm, Sweden.
| | - Jianchun Yu
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
27
|
Gu T, Tian X, Wang Y, Yang W, Li W, Song M, Zhao R, Wang M, Gao Q, Li T, Zhang C, Kundu JK, Liu K, Dong Z, Lee MH. Repurposing pentamidine for cancer immunotherapy by targeting the PD1/PD-L1 immune checkpoint. Front Immunol 2023; 14:1145028. [PMID: 37205112 PMCID: PMC10185823 DOI: 10.3389/fimmu.2023.1145028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/10/2023] [Indexed: 05/21/2023] Open
Abstract
Immunotherapy has emerged as an effective therapeutic approach to several cancer types. The reinvigoration of tumor-infiltrating lymphocyte-mediated immune responses via the blockade of immune checkpoint markers, such as program cell death-1 (PD-1) or its cognate ligand PD-L1, has been the basis for developing clinically effective anticancer therapies. We identified pentamidine, an FDA-approved antimicrobial agent, as a small-molecule antagonist of PD-L1. Pentamidine enhanced T-cell-mediated cytotoxicity against various cancer cells in vitro by increasing the secretion of IFN-γ, TNF-α, perforin, and granzyme B in the culture medium. Pentamidine promoted T-cell activation by blocking the PD-1/PD-L1 interaction. In vivo administration of pentamidine attenuated the tumor growth and prolonged the survival of tumor-bearing mice in PD-L1 humanized murine tumor cell allograft models. Histological analysis of tumor tissues showed an increased number of tumor-infiltrating lymphocytes in tissues derived from pentamidine-treated mice. In summary, our study suggests that pentamidine holds the potential to be repurposed as a novel PD-L1 antagonist that may overcome the limitations of monoclonal antibody therapy and can emerge as a small molecule cancer immunotherapy.
Collapse
Affiliation(s)
- Tingxuan Gu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Xueli Tian
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Yuanyuan Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Wenqian Yang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Wenwen Li
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
| | - Mengqiu Song
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Ran Zhao
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Mengqiao Wang
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
| | - Quanli Gao
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Tiepeng Li
- Department of Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Chengjuan Zhang
- Department of Immunology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Joydeb Kumar Kundu
- Li Ka Shing Applied Virology Institute, University of Alberta, Edmonton, AB, Canada
| | - Kangdong Liu
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China
| | - Zigang Dong
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- *Correspondence: Zigang Dong, ; Mee-Hyun Lee,
| | - Mee-Hyun Lee
- Department of Pathophysiology, School of Basic Medical Sciences, Academy of Medical Science, College of Medicine, Zhengzhou University, Zhengzhou, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, China
- Henan Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University, Zhengzhou, China
- College of Korean Medicine, Dongshin University, Naju, Republic of Korea
- *Correspondence: Zigang Dong, ; Mee-Hyun Lee,
| |
Collapse
|
28
|
Ghaedrahmati F, Esmaeil N, Abbaspour M. Targeting immune checkpoints: how to use natural killer cells for fighting against solid tumors. Cancer Commun (Lond) 2022; 43:177-213. [PMID: 36585761 PMCID: PMC9926962 DOI: 10.1002/cac2.12394] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/08/2022] [Accepted: 11/15/2022] [Indexed: 01/01/2023] Open
Abstract
Natural killer (NK) cells are unique innate immune cells that mediate anti-viral and anti-tumor responses. Thus, they might hold great potential for cancer immunotherapy. NK cell adoptive immunotherapy in humans has shown modest efficacy. In particular, it has failed to demonstrate therapeutic efficiency in the treatment of solid tumors, possibly due in part to the immunosuppressive tumor microenvironment (TME), which reduces NK cell immunotherapy's efficiencies. It is known that immune checkpoints play a prominent role in creating an immunosuppressive TME, leading to NK cell exhaustion and tumor immune escape. Therefore, NK cells must be reversed from their dysfunctional status and increased in their effector roles in order to improve the efficiency of cancer immunotherapy. Blockade of immune checkpoints can not only rescue NK cells from exhaustion but also augment their robust anti-tumor activity. In this review, we discussed immune checkpoint blockade strategies with a focus on chimeric antigen receptor (CAR)-NK cells to redirect NK cells to cancer cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Farhoodeh Ghaedrahmati
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Nafiseh Esmaeil
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran,Research Institute for Primordial Prevention of Non‐Communicable DiseaseIsfahan University of Medical SciencesIsfahanIran
| | - Maryam Abbaspour
- Department of Pharmaceutical BiotechnologyFaculty of PharmacyIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
29
|
Li Y, Luo Z, Wang X, Zhang S, Hei H, Qin J. Design of new drugs for medullary thyroid carcinoma. Front Oncol 2022; 12:993725. [PMID: 36544713 PMCID: PMC9760674 DOI: 10.3389/fonc.2022.993725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 11/18/2022] [Indexed: 12/08/2022] Open
Abstract
Medullary thyroid carcinoma (MTC) is one of the common malignant endocrine tumors, which seriously affects human health. Although surgical resection offers a potentially curative therapeutic option to some MTC patients, most patients do not benefit from it due to the difficulty to access the tumors and tumor metastasis. The survival rate of MTC patients has improved with the recent advances in the research, which has improved our understanding of the molecular mechanism underlying MTC and enabled the development and approval of novel targeted drugs. In this article, we reviewed the molecular mechanisms related to MTC progression and the principle for the design of molecular targeted drugs, and proposed some future directions for prospective studies exploring targeted drugs for MTC.
Collapse
Affiliation(s)
- Yanqing Li
- Department of Thyroid and Neck, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China,The Medical School of Zhengzhou University, Zhengzhou, China
| | - Ziyu Luo
- Department of Thyroid and Neck, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China,The Medical School of Zhengzhou University, Zhengzhou, China
| | - Xinxing Wang
- Department of Pain and Rehabilitation and Palliative Medicine, Henan Cancer Hospital, Zhengzhou, China,*Correspondence: Songtao Zhang, ; Hu Hei, ; Jianwu Qin, ; Xinxing Wang,
| | - Songtao Zhang
- Department of Thyroid and Neck, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China,The Medical School of Zhengzhou University, Zhengzhou, China,*Correspondence: Songtao Zhang, ; Hu Hei, ; Jianwu Qin, ; Xinxing Wang,
| | - Hu Hei
- Department of Thyroid and Neck, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China,The Medical School of Zhengzhou University, Zhengzhou, China,*Correspondence: Songtao Zhang, ; Hu Hei, ; Jianwu Qin, ; Xinxing Wang,
| | - Jianwu Qin
- Department of Thyroid and Neck, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China,The Medical School of Zhengzhou University, Zhengzhou, China,*Correspondence: Songtao Zhang, ; Hu Hei, ; Jianwu Qin, ; Xinxing Wang,
| |
Collapse
|
30
|
Joseph J, Rahmani B, Cole Y, Puttagunta N, Lin E, Khan ZK, Jain P. Can Soluble Immune Checkpoint Molecules on Exosomes Mediate Inflammation? J Neuroimmune Pharmacol 2022; 17:381-397. [PMID: 34697721 PMCID: PMC10128092 DOI: 10.1007/s11481-021-10018-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/25/2021] [Indexed: 01/13/2023]
Abstract
Immune checkpoints (ICPs) are major co-signaling pathways that trigger effector functions in immune cells, with isoforms that are either membrane bound, engaging in direct cell to cell activation locally, or soluble, acting at distant sites by circulating freely or potentially via extracellular vesicles (EVs). Exosomes are small EVs secreted by a variety of cells carrying various proteins and nucleic acids. They are distributed extensively through biological fluids and have major impacts on infectious diseases, cancer, and neuroinflammation. Similarly, ICPs play key roles in a variety of disease conditions and have been extensively utilized as a prognostic tool for various cancers. Herein, we explored if the association between exosomes and ICPs could be a significant contributor of inflammation, particularly in the setting of cancer, neuroinflammation and viral infections, wherein the up regulation in both exosomal proteins and ICPs correlate with immunosuppressive effects. The detailed literature review of existing data highlights the significance and complexity of these two important pathways in mediating cancer and potentiating neuroinflammation via modulating overall immune response. Cells increasingly secret exosomes in response to intracellular signals from invading pathogens or cancerous transformations. These exosomes can carry a variety of cargo including proteins, nucleic acids, cytokines, and receptors/ligands that have functional consequences on recipient cells. Illustration generated using BioRender software.
Collapse
Affiliation(s)
- Julie Joseph
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Benjamin Rahmani
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Yonesha Cole
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Neha Puttagunta
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Edward Lin
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Zafar K Khan
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA
| | - Pooja Jain
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA. .,Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 West Queen Lane, Philadelphia, PA, 19129, USA.
| |
Collapse
|
31
|
Mutant p53 gain of function mediates cancer immune escape that is counteracted by APR-246. Br J Cancer 2022; 127:2060-2071. [PMID: 36138076 PMCID: PMC9681866 DOI: 10.1038/s41416-022-01971-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 08/05/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND p53 mutants contribute to the chronic inflammatory tumour microenvironment (TME). In this study, we address the mechanism of how p53 mutants lead to chronic inflammation in tumours and how to transform it to restore cancer immune surveillance. METHODS Our analysis of RNA-seq data from The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) project revealed that mutant p53 (mtp53) cancers correlated with chronic inflammation. We used cell-based assays and a mouse model to discover a novel gain of function of mtp53 and the effect of the mtp53 reactivating compound APR-246 on the anti-tumour immune response. RESULTS We found that tumour samples from patients with breast carcinoma carrying mtp53 showed elevated Interferon (IFN) signalling, Tumour Inflammation Signature (TIS) score and infiltration of CD8+ T cells compared to wild type p53 (wtp53) tumours. We showed that the expression of IFN and immune checkpoints were elevated in tumour cells in a mtp53-dependent manner, suggesting a novel gain of function. Restoration of wt function to mtp53 by APR-246 induced the expression of endogenous retroviruses, IFN signalling and repressed immune checkpoints. Moreover, APR-246 promoted CD4+ T cells infiltration and IFN signalling and prevented CD8+ T cells exhaustion within the TME in vivo. CONCLUSIONS Breast carcinomas with mtp53 displayed enhanced inflammation. APR-246 boosted the interferon response or represses immune checkpoints in p53 mutant tumour cells, and restores cancer immune surveillance in vivo.
Collapse
|
32
|
Lanier OL, Pérez-Herrero E, Andrea APD, Bahrami K, Lee E, Ward DM, Ayala-Suárez N, Rodríguez-Méndez SM, Peppas NA. Immunotherapy approaches for hematological cancers. iScience 2022; 25:105326. [PMID: 36325064 PMCID: PMC9619355 DOI: 10.1016/j.isci.2022.105326] [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: 12/24/2022] Open
Abstract
Hematological cancers such as leukemia, lymphoma, and multiple myeloma have traditionally been treated with chemo and radiotherapy approaches. Introduction of immunotherapies for treatment of these diseases has led to patient remissions that would not have been possible with traditional approaches. In this critical review we identify main disease characteristics, symptoms, and current treatment options. Five common immunotherapies, namely checkpoint inhibitors, vaccines, cell-based therapies, antibodies, and oncolytic viruses, are described, and their applications in hematological cancers are critically discussed.
Collapse
Affiliation(s)
- Olivia L. Lanier
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Edgar Pérez-Herrero
- Departamento de Ingeniería Química y Tecnología Farmacéutica, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
- Instituto Universitario de Tecnologías Biomédicas, Universidad de La Laguna, La Laguna, 38200 Tenerife, Spain
| | - Abielle P. D.’ Andrea
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Kiana Bahrami
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Elaine Lee
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Deidra M. Ward
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Nilaya Ayala-Suárez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Sheyla M. Rodríguez-Méndez
- Instituto Universitario de Bio-Orgánica Antonio González, Universidad de La Laguna, La Laguna, 38206 Tenerife, Spain
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA
- Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
33
|
Minott JA, van Vloten JP, Yates JGE, Chan L, Wood GA, Viloria-Petit AM, Karimi K, Petrik JJ, Wootton SK, Bridle BW. Multiplex flow cytometry-based assay for quantifying tumor- and virus-associated antibodies induced by immunotherapies. Front Immunol 2022; 13:1038340. [PMID: 36466867 PMCID: PMC9708883 DOI: 10.3389/fimmu.2022.1038340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/27/2022] [Indexed: 03/22/2024] Open
Abstract
Novel immunotherapies continue to be developed and tested for application against a plethora of diseases. The clinical translation of immunotherapies requires an understanding of their mechanisms. The contributions of antibodies in driving long-term responses following immunotherapies continue to be revealed given their diverse effector functions. Developing an in-depth understanding of the role of antibodies in treatment efficacy is required to optimize immunotherapies and improve the chance of successfully translating them into the clinic. However, analyses of antibody responses can be challenging in the context of antigen-agnostic immunotherapies, particularly in the context of cancers that lack pre-defined target antigens. As such, robust methods are needed to evaluate the capacity of a given immunotherapy to induce beneficial antibody responses, and to identify any therapy-limiting antibodies. We previously developed a comprehensive method for detecting antibody responses induced by antigen-agnostic immunotherapies for application in pre-clinical models of vaccinology and cancer therapy. Here, we extend this method to a high-throughput, flow cytometry-based assay able to identify and quantify isotype-specific virus- and tumor-associated antibody responses induced by immunotherapies using small sample volumes with rapid speed and high sensitivity. This method provides a valuable and flexible protocol for investigating antibody responses induced by immunotherapies, which researchers can use to expand their analyses and optimize their own treatment regimens.
Collapse
Affiliation(s)
- Jessica A. Minott
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | | | - Jacob G. E. Yates
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Lily Chan
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Geoffrey A. Wood
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | | | - Khalil Karimi
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - James J. Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON, Canada
- ImmunoCeutica Inc., Cambridge, ON, Canada
| |
Collapse
|
34
|
Diep YN, Kim TJ, Cho H, Lee LP. Nanomedicine for advanced cancer immunotherapy. J Control Release 2022; 351:1017-1037. [DOI: 10.1016/j.jconrel.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/30/2022] [Accepted: 10/01/2022] [Indexed: 11/09/2022]
|
35
|
Anderson TS, Wooster AL, Piersall SL, Okpalanwaka IF, Lowe DB. Disrupting cancer angiogenesis and immune checkpoint networks for improved tumor immunity. Semin Cancer Biol 2022; 86:981-996. [PMID: 35149179 PMCID: PMC9357867 DOI: 10.1016/j.semcancer.2022.02.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 02/05/2022] [Indexed: 01/27/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have advanced the field of cancer immunotherapy in patients by sustaining effector immune cell activity within the tumor microenvironment. However, the approach in general is still faced with issues related to ICI response duration/resistance, treatment eligibility, and safety, which indicates a need for further refinements. As immune checkpoint upregulation is inextricably linked to cancer-induced angiogenesis, newer clinical efforts have demonstrated the feasibility of disrupting both tumor-promoting networks to mediate enhanced immune-driven protection. This review focuses on such key evidence stipulating the necessity of co-applying ICI and anti-angiogenic strategies in cancer patients, with particular interest in highlighting newer engineered antibody approaches that may provide theoretically superior multi-pronged and safe therapeutic combinations.
Collapse
Affiliation(s)
- Trevor S Anderson
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Amanda L Wooster
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Savanna L Piersall
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Izuchukwu F Okpalanwaka
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States
| | - Devin B Lowe
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, TX, 79601, United States.
| |
Collapse
|
36
|
Sauer N, Szlasa W, Jonderko L, Oślizło M, Kunachowicz D, Kulbacka J, Karłowicz-Bodalska K. LAG-3 as a Potent Target for Novel Anticancer Therapies of a Wide Range of Tumors. Int J Mol Sci 2022; 23:9958. [PMID: 36077354 PMCID: PMC9456311 DOI: 10.3390/ijms23179958] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/22/2022] [Accepted: 08/29/2022] [Indexed: 12/20/2022] Open
Abstract
LAG-3 (Lymphocyte activation gene 3) protein is a checkpoint receptor that interacts with LSEC-tin, Galectin-3 and FGL1. This interaction leads to reduced production of IL-2 and IFN-γ. LAG-3 is widely expressed in different tumor types and modulates the tumor microenvironment through immunosuppressive effects. Differential expression in various tumor types influences patient prognosis, which is often associated with coexpression with immune checkpoint inhibitors, such as TIM-3, PD-1 and CTLA-4. Here, we discuss expression profiles in different tumor types. To date, many clinical trials have been conducted using LAG-3 inhibitors, which can be divided into anti-LAG-3 monoclonal antibodies, anti-LAG-3 bispecifics and soluble LAG-3-Ig fusion proteins. LAG-3 inhibitors supress T-cell proliferation and activation by disallowing for the interaction between LAG-3 to MHC-II. The process enhances anti-tumor immune response. In this paper, we will review the current state of knowledge on the structure, function and expression of LAG-3 in various types of cancer, as well as its correlation with overall prognosis, involvement in cell-based therapies and experimental medicine. We will consider the role of compounds targeting LAG-3 in clinical trials both as monotherapy and in combination, which will provide data relating to the efficacy and safety of proposed drug candidates.
Collapse
Affiliation(s)
- Natalia Sauer
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, 50-367 Wroclaw, Poland
| | - Laura Jonderko
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | | | | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | |
Collapse
|
37
|
Hassanian H, Asadzadeh Z, Baghbanzadeh A, Derakhshani A, Dufour A, Rostami Khosroshahi N, Najafi S, Brunetti O, Silvestris N, Baradaran B. The expression pattern of Immune checkpoints after chemo/radiotherapy in the tumor microenvironment. Front Immunol 2022; 13:938063. [PMID: 35967381 PMCID: PMC9367471 DOI: 10.3389/fimmu.2022.938063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
As a disease with the highest disease-associated burden worldwide, cancer has been the main subject of a considerable proportion of medical research in recent years, intending to find more effective therapeutic approaches with fewer side effects. Combining conventional methods with newer biologically based treatments such as immunotherapy can be a promising approach to treating different tumors. The concept of "cancer immunoediting" that occurs in the field of the tumor microenvironment (TME) is the aspect of cancer therapy that has not been at the center of attention. One group of the role players of the so-called immunoediting process are the immune checkpoint molecules that exert either co-stimulatory or co-inhibitory effects in the anti-tumor immunity of the host. It involves alterations in a wide variety of immunologic pathways. Recent studies have proven that conventional cancer therapies, such as chemotherapy, radiotherapy, or a combination of them, i.e., chemoradiotherapy, alter the "immune compartment" of the TME. The mentioned changes encompass a wide range of variations, including the changes in the density and immunologic type of the tumor-infiltrating lymphocytes (TILs) and the alterations in the expression patterns of the different immune checkpoints. These rearrangements can have either anti-tumor immunity empowering or immune attenuating sequels. Thus, recognizing the consequences of various chemo(radio)therapeutic regimens in the TME seems to be of great significance in the evolution of therapeutic approaches. Therefore, the present review intends to summarize how chemo(radio)therapy affects the TME and specifically some of the most important, well-known immune checkpoints' expressions according to the recent studies in this field.
Collapse
Affiliation(s)
- Hamidreza Hassanian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Departments of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | | | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi” University of Messina, Messina, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
Staros R, Michalak A, Rusinek K, Mucha K, Pojda Z, Zagożdżon R. Perspectives for 3D-Bioprinting in Modeling of Tumor Immune Evasion. Cancers (Basel) 2022; 14:cancers14133126. [PMID: 35804898 PMCID: PMC9265021 DOI: 10.3390/cancers14133126] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
In a living organism, cancer cells function in a specific microenvironment, where they exchange numerous physical and biochemical cues with other cells and the surrounding extracellular matrix (ECM). Immune evasion is a clinically relevant phenomenon, in which cancer cells are able to direct this interchange of signals against the immune effector cells and to generate an immunosuppressive environment favoring their own survival. A proper understanding of this phenomenon is substantial for generating more successful anticancer therapies. However, classical cell culture systems are unable to sufficiently recapture the dynamic nature and complexity of the tumor microenvironment (TME) to be of satisfactory use for comprehensive studies on mechanisms of tumor immune evasion. In turn, 3D-bioprinting is a rapidly evolving manufacture technique, in which it is possible to generate finely detailed structures comprised of multiple cell types and biomaterials serving as ECM-analogues. In this review, we focus on currently used 3D-bioprinting techniques, their applications in the TME research, and potential uses of 3D-bioprinting in modeling of tumor immune evasion and response to immunotherapies.
Collapse
Affiliation(s)
- Rafał Staros
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Agata Michalak
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Kinga Rusinek
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Krzysztof Mucha
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
| | - Zygmunt Pojda
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
| | - Radosław Zagożdżon
- Department of Immunology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-006 Warsaw, Poland; (R.S.); (K.M.)
- Department of Regenerative Medicine, Maria Sklodowska-Curie National Institute of Oncology, 02-781 Warsaw, Poland; (A.M.); (K.R.); (Z.P.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-006 Warsaw, Poland
- Correspondence: ; Tel.: +48-22-502-14-72; Fax: +48-22-502-21-59
| |
Collapse
|
39
|
Effects of Nivolumab and Ipilimumab on the suppression of cisplatin resistant small cell lung cancer cells. Invest New Drugs 2022; 40:709-717. [PMID: 35438354 DOI: 10.1007/s10637-022-01243-5] [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: 02/06/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Small cell lung cancer (SCLC) accounts for nearly 10-15% of all lung cancer cases. Although many chemotherapy drugs, such as cisplatin and etoposide, were approved as primary therapy for SCLC patients, the prognosis is poor. In this study, we aimed to explore novel therapeutic strategy against SCLC. METHODS Two SCLC cell lines, LTEP-P and LTEP-P/DDP1.0, were treated with cisplatin, in the absence or presence of Nivolumab + Ipilimumab combination, and the cell viability was measured. Tumor size and mouse survival rate were examined upon different drug treatments. Protein levels of PD-1 and CTLA4 were detected in normal and SCLC cells by Western blot. Cellular cytotoxicity induced by T lymphocytes was measured by thymidine incorporation assay. Tumor infiltrated T cell populations from LTEP-P and LTEP/DDP1.0 tumor-bearing mice were analyzed by flow cytometry. RESULTS LTEP-P cells, but not LTEP/DDP1.0 cells, exhibited decreased cell viability upon cisplatin, Nivolumab and Ipilimumab combinational treatment. T lymphocytes significantly inhibited the growth of LTEP-P cells in the presence of nivolumab and ipilimumab. The combinational therapy improved survival rate and inhibited tumor growth in LTEP-P tumor-bearing mice, but showed no effect on LTEP/DDP1.0 tumor-bearing mice. Nivolumab and Ipilimumab synergized with cisplatin in increasing CD8 + and CD4 + T cell population, while decreasing Treg population in LTEP-P tumor-bearing mice. CONCLUSIONS The combinational therapy by cisplatin, Nivolumab and Ipilimumab could be an effective strategy against LTEP-P cells, accompanied with increased cytotoxic T cell populations, but has no significant effect against DDP-resistant lung adenocarcinoma cells.
Collapse
|
40
|
Karami Fath M, Babakhaniyan K, Zokaei M, Yaghoubian A, Akbari S, Khorsandi M, Soofi A, Nabi-Afjadi M, Zalpoor H, Jalalifar F, Azargoonjahromi A, Payandeh Z, Alagheband Bahrami A. Anti-cancer peptide-based therapeutic strategies in solid tumors. Cell Mol Biol Lett 2022; 27:33. [PMID: 35397496 PMCID: PMC8994312 DOI: 10.1186/s11658-022-00332-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Nowadays, conventional medical treatments such as surgery, radiotherapy, and chemotherapy cannot cure all types of cancer. A promising approach to treat solid tumors is the use of tumor-targeting peptides to deliver drugs or active agents selectively. RESULT Introducing beneficial therapeutic approaches, such as therapeutic peptides and their varied methods of action against tumor cells, can aid researchers in the discovery of novel peptides for cancer treatment. The biomedical applications of therapeutic peptides are highly interesting. These peptides, owing to their high selectivity, specificity, small dimensions, high biocompatibility, and easy modification, provide good opportunities for targeted drug delivery. In recent years, peptides have shown considerable promise as therapeutics or targeting ligands in cancer research and nanotechnology. CONCLUSION This study reviews a variety of therapeutic peptides and targeting ligands in cancer therapy. Initially, three types of tumor-homing and cell-penetrating peptides (CPPs) are described, and then their applications in breast, glioma, colorectal, and melanoma cancer research are discussed.
Collapse
Affiliation(s)
- Mohsen Karami Fath
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Kimiya Babakhaniyan
- Department of Medical Surgical Nursing, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Zokaei
- Department of Food Science and Technology, Faculty of Nutrition Science, Food Science and Technology/National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Veterinary Medicine, Beyza Branch, Islamic Azad University, Beyza, Iran
| | - Azadeh Yaghoubian
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Akbari
- Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahdieh Khorsandi
- Department of Biotechnology, Faculty of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Asma Soofi
- Department of Physical Chemistry, School of Chemistry, College of Sciences, University of Tehran, Tehran, Iran
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of biological science, Tarbiat Modares University, Tehran, Iran
| | - Hamidreza Zalpoor
- American Association of Kidney Patients, Tampa, FL USA
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Network of Immunity in Infection, Malignancy & Autoimmunity (NIIMA), Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Fateme Jalalifar
- School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | | | - Zahra Payandeh
- Department Medical Biochemistry and Biophysics, Division Medical Inflammation Research, Karolinska Institute, Stockholm, Sweden
| | - Armina Alagheband Bahrami
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
41
|
Muhsin A, Rangel R, Vien L, Bover L. Monoclonal Antibodies Generation: Updates and Protocols on Hybridoma Technology. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2435:73-93. [PMID: 34993940 DOI: 10.1007/978-1-0716-2014-4_6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Since its inception in 1975, the hybridoma technology revolutionized science and medicine, facilitating discoveries in almost any field from the laboratory to the clinic. Many technological advancements have been developed since then, to create these "magical bullets." Phage and yeast display libraries expressing the variable heavy and light domains of antibodies, single B-cell cloning from immunized animals of different species including humans or in silico approaches, all have rendered a myriad of newly developed antibodies or improved design of existing ones. However, still the majority of these antibodies or their recombinant versions are from hybridoma origin, a preferred methodology that trespass species barriers, due to the preservation of the natural functions of immune cells in producing the humoral response: antigen specific immunoglobulins. Remarkably, this methodology can be reproduced in small laboratories without the need of sophisticate equipment. In this chapter, we will describe the most recent methods utilized by our Monoclonal Antibodies Core Facility at the University of Texas-M.D. Anderson Cancer Center. During the last 10 years, the methods, techniques, and expertise implemented in our core had generated more than 350 antibodies for various applications.
Collapse
Affiliation(s)
- Ahmed Muhsin
- Department of Immunology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA.,Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Roberto Rangel
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Long Vien
- Department of Immunology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA
| | - Laura Bover
- Department of Immunology, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA. .,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX, USA.
| |
Collapse
|
42
|
Unraveling the Expression Patterns of Immune Checkpoints Identifies New Subtypes and Emerging Therapeutic Indicators in Lung Adenocarcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3583985. [PMID: 35178154 PMCID: PMC8843963 DOI: 10.1155/2022/3583985] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 01/06/2022] [Indexed: 12/16/2022]
Abstract
Immune checkpoint genes (ICGs) play pivotal roles in tumor immune microenvironment (TIME), and thus, targeting them represents a promising strategy for cancer immunotherapy. However, the genetic landscape of ICGs in lung adenocarcinoma (LUAD) is still unknown. Herein, we comprehensively evaluated the ICG expression profiles of 1439 LUAD samples and linked ICG expression patterns with infiltration of immune cells, clinical features, and response to immune checkpoint blockade (ICB). The ICGscore was developed to quantify ICG expression patterns of individual patient by principal component analysis algorithms. Three distinct ICG expression patterns and three ICG-related genomic clusters were determined, which were implicated in different clinical outcomes, level of immune infiltrates, and biological process. LUAD patients were subdivided into high- and low-ICGscore subgroups. Patients with higher ICGscore were characterized by favorable survival outcomes, increased immune cell infiltration, and enhanced expression of ICGs. Further analysis revealed that lower ICGscore was associated with greater tumor mutation loads and higher mutation rates of TTN, KEAP1, and ZFHX4. High ICGscore has the potential to be a robust indicator in clinical benefit of immunotherapy. Taken together, unraveling the ICG expression patterns will advance our understanding of heterogeneity of TIME and guides more effective immunotherapeutic strategies in LUAD.
Collapse
|
43
|
Charpentier M, Dupré E, Fortun A, Briand F, Maillasson M, Com E, Pineau C, Labarrière N, Rabu C, Lang F. hnRNP-A1 binds to the IRES of MELOE-1 antigen to promote MELOE-1 translation in stressed melanoma cells. Mol Oncol 2022; 16:594-606. [PMID: 34418284 PMCID: PMC8807352 DOI: 10.1002/1878-0261.13088] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/05/2021] [Accepted: 08/20/2021] [Indexed: 12/29/2022] Open
Abstract
The major challenge in antigen-specific immunotherapy of cancer is to select the most relevant tumor antigens to target. To this aim, understanding their mode of expression by tumor cells is critical. We previously identified a melanoma-specific antigen, melanoma-overexpressed antigen 1 (MELOE-1)-coded for by a long noncoding RNA-whose internal ribosomal entry sequence (IRES)-dependent translation is restricted to tumor cells. This restricted expression is associated with the presence of a broad-specific T-cell repertoire that is involved in tumor immunosurveillance in melanoma patients. In the present work, we explored the translation control of MELOE-1 and provide evidence that heterogeneous nuclear ribonucleoprotein A1 (hnRNP-A1) binds to the MELOE-1 IRES and acts as an IRES trans-activating factor (ITAF) to promote the translation of MELOE-1 in melanoma cells. In addition, we showed that endoplasmic reticulum (ER) stress induced by thapsigargin, which promotes hnRNP-A1 cytoplasmic translocation, enhances MELOE-1 translation and recognition of melanoma cells by a MELOE-1-specific T-cell clone. These findings suggest that pharmacological stimulation of stress pathways may enhance the efficacy of immunotherapies targeting stress-induced tumor antigens such as MELOE-1.
Collapse
Affiliation(s)
| | - Emilie Dupré
- InsermLabEx IGOCRCINAUniversité de NantesNantesFrance
| | - Agnès Fortun
- InsermLabEx IGOCRCINAUniversité de NantesNantesFrance
| | | | - Mike Maillasson
- InsermLabEx IGOCRCINAUniversité de NantesNantesFrance
- InsermCNRSSFR SantéInserm UMS 016CNRS UMS 3556Université de NantesNantesFrance
| | - Emmanuelle Com
- InsermEHESPIrset (Institut de recherche en santé, environnement et travail) – UMR‐S 1085Univ RennesRennesFrance
- ProtimBiosit – UMS 3480US‐S 018Univ RennesRennesFrance
| | - Charles Pineau
- InsermEHESPIrset (Institut de recherche en santé, environnement et travail) – UMR‐S 1085Univ RennesRennesFrance
- ProtimBiosit – UMS 3480US‐S 018Univ RennesRennesFrance
| | | | | | - François Lang
- InsermLabEx IGOCRCINAUniversité de NantesNantesFrance
| |
Collapse
|
44
|
Bajor M, Graczyk-Jarzynka A, Marhelava K, Burdzinska A, Muchowicz A, Goral A, Zhylko A, Soroczynska K, Retecki K, Krawczyk M, Klopotowska M, Pilch Z, Paczek L, Malmberg KJ, Wälchli S, Winiarska M, Zagozdzon R. PD-L1 CAR effector cells induce self-amplifying cytotoxic effects against target cells. J Immunother Cancer 2022; 10:jitc-2021-002500. [PMID: 35078921 PMCID: PMC8796262 DOI: 10.1136/jitc-2021-002500] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 12/21/2022] Open
Abstract
BackgroundImmune checkpoint inhibitors and chimeric antigen receptor (CAR)-based therapies have transformed cancer treatment. Recently, combining these approaches into a strategy of PD-L1-targeted CAR has been proposed to target PD-L1high tumors. Our study provides new information on the efficacy of such an approach against PD-L1low targets.MethodsNew atezolizumab-based PD-L1-targeted CAR was generated and introduced into T, NK, or NK-92 cells. Breast cancer MDA-MB-231 and MCF-7 cell lines or non-malignant cells (HEK293T, HMEC, MCF-10A, or BM-MSC) were used as targets to assess the reactivity or cytotoxic activity of the PD-L1–CAR-bearing immune effector cells. Stimulation with IFNγ or with supernatants from activated CAR T cells were used to induce upregulation of PD-L1 molecule expression on the target cells. HER2–CAR T cells were used for combination with PD-L1–CAR T cells against MCF-7 cells.ResultsPD-L1–CAR effector cells responded vigorously with degranulation and cytokine production to PD-L1high MDA-MB-231 cells, but not to PD-L1low MCF-7 cells. However, in long-term killing assays, both MDA-MB-231 and MCF-7 cells were eliminated by the PD-L1–CAR cells, although with a delay in the case of PD-L1low MCF-7 cells. Notably, the coculture of MCF-7 cells with activated PD-L1–CAR cells led to bystander induction of PD-L1 expression on MCF-7 cells and to the unique self-amplifying effect of the PD-L1–CAR cells. Accordingly, PD-L1–CAR T cells were active not only against MDA-MD-231 and MCF-7-PD-L1 but also against MCF-7-pLVX cells in tumor xenograft models. Importantly, we have also observed potent cytotoxic effects of PD-L1–CAR cells against non-malignant MCF-10A, HMEC, and BM-MSC cells, but not against HEK293T cells that initially did not express PD-L1 and were unresponsive to the stimulation . Finally, we have observed that HER-2–CAR T cells stimulate PD-L1 expression on MCF-7 cells and therefore accelerate the functionality of PD-L1–CAR T cells when used in combination.ConclusionsIn summary, our studies show that CAR-effector cells trigger the expression of PD-L1 on target cells, which in case of PD-L1–CAR results in the unique self-amplification phenomenon. This self-amplifying effect could be responsible for the enhanced cytotoxicity of PD-L1–CAR T cells against both malignant and non-malignant cells and implies extensive caution in introducing PD-L1–CAR strategy into clinical studies.
Collapse
Affiliation(s)
- Malgorzata Bajor
- Department of Clinical Immunology, Medical University of Warsaw, Warszawa, Mazowieckie, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Graczyk-Jarzynka
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
| | - Katsiaryna Marhelava
- Department of Clinical Immunology, Medical University of Warsaw, Warszawa, Mazowieckie, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
- Laboratory for Cellular and Genetic Therapies, Medical University of Warsaw, Warsaw, Poland
| | - Anna Burdzinska
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warszawa, Poland
| | - Angelika Muchowicz
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
| | - Agnieszka Goral
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
| | - Andriy Zhylko
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
- Doctoral School, Medical University of Warsaw, Warsaw, Poland
| | | | - Kuba Retecki
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
| | - Marta Krawczyk
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
- Laboratory for Cellular and Genetic Therapies, Medical University of Warsaw, Warsaw, Poland
- Doctoral School of Translational Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Marta Klopotowska
- Department of Clinical Immunology, Medical University of Warsaw, Warszawa, Mazowieckie, Poland
- Laboratory of Immunology, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Zofia Pilch
- Department of Immunology, Medical University of Warsaw, Warszawa, Poland
| | - Leszek Paczek
- Department of Immunology, Transplantology and Internal Diseases, Medical University of Warsaw, Warszawa, Poland
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Sébastien Wälchli
- Department of Cellular Therapy, Oslo University Hospital, Oslo, Norway
| | | | - Radoslaw Zagozdzon
- Department of Clinical Immunology, Medical University of Warsaw, Warszawa, Mazowieckie, Poland
- Laboratory for Cellular and Genetic Therapies, Medical University of Warsaw, Warsaw, Poland
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Department of Regenerative Medicine, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
45
|
Nadesh R, Menon KN, Biswas L, Mony U, Subramania Iyer K, Vijayaraghavan S, Nambiar A, Nair S. Adipose derived mesenchymal stem cell secretome formulation as a biotherapeutic to inhibit growth of drug resistant triple negative breast cancer. Sci Rep 2021; 11:23435. [PMID: 34873206 PMCID: PMC8648896 DOI: 10.1038/s41598-021-01878-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 10/22/2021] [Indexed: 12/30/2022] Open
Abstract
In the present study, a protocol was developed for processing of human adipose derived mesenchymal stem cell secretome formulation of varying concentration. Its molecular composition was evaluated, and its effectiveness in vitro using breast cancer cell lines, and in vivo in a nude mice breast cancer model was studied to determine its role in suppressing triple negative breast cancer in a dose dependent manner. Because the secretome could have value as an add-on therapy along with a current drug, the effectiveness of the secretome both in monotherapy and in combination therapy along with paclitaxel was evaluated. The results showed significant cell kill when exposed to the secretome above 20 mg/ml at which concentration there was no toxicity to normal cells. 70 mg/ml of SF showed 90 ± 10% apoptosis and significant decrease in CD44+/CD24−, MDR1+ and PDL-1+ cancer cells. In vivo, the tumor showed no growth after daily intra tumor injections at 50 mg/ml and 100 mg/ml doses whereas substantial tumor growth occurred after saline intra tumor injection. The study concludes that SF is a potential biotherapeutic for breast cancer and could be used initially as an add-on therapy to other standard of care to provide improved efficacy without other adverse effects.
Collapse
Affiliation(s)
- Ragima Nadesh
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Krishnakumar N Menon
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Lalitha Biswas
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Ullas Mony
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - K Subramania Iyer
- Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Sundeep Vijayaraghavan
- Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Ajit Nambiar
- Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Shantikumar Nair
- Amrita Centre for Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| |
Collapse
|
46
|
The Immune Landscape of Breast Cancer: Strategies for Overcoming Immunotherapy Resistance. Cancers (Basel) 2021; 13:cancers13236012. [PMID: 34885122 PMCID: PMC8657247 DOI: 10.3390/cancers13236012] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/25/2021] [Accepted: 11/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Immunotherapy is a rapidly advancing field in breast cancer treatment, however, it encounters many obstacles that leave open gateways for breast cancer cells to resist novel immunotherapies. It is believed that the tumor microenvironment consisting of cancer, stromal, and immune cells as well as a plethora of tumor-promoting soluble factors, is responsible for the failure of therapeutic strategies in cancer, including breast tumors. Therefore, an in-depth understanding of key barriers to effective immunotherapy, focusing the research efforts on harnessing the power of the immune system, and thus, developing new strategies to overcome the resistance may contribute significantly to increase breast cancer patient survival. In this review, we discuss the latest reports regarding the strategies rendering the immunosuppressive tumor microenvironment more sensitive to immunotherapy in breast cancers, HER2-positive and triple-negative types of breast cancer, which are attractive from an immunotherapeutic point of view. Abstract Breast cancer (BC) has traditionally been considered to be not inherently immunogenic and insufficiently represented by immune cell infiltrates. Therefore, for a long time, it was thought that the immunotherapies targeting this type of cancer and its microenvironment were not justified and would not bring benefits for breast cancer patients. Nevertheless, to date, a considerable number of reports have indicated tumor-infiltrating lymphocytes (TILs) as a prognostic and clinically relevant biomarker in breast cancer. A high TILs expression has been demonstrated in primary tumors, of both, HER2-positive BC and triple-negative (TNBC), of patients before treatment, as well as after treatment with adjuvant and neoadjuvant chemotherapy. Another milestone was reached in advanced TNBC immunotherapy with the help of the immune checkpoint inhibitors directed against the PD-L1 molecule. Although those findings, together with the recent developments in chimeric antigen receptor T cell therapies, show immense promise for significant advancements in breast cancer treatments, there are still various obstacles to the optimal activity of immunotherapeutics in BC treatment. Of these, the immunosuppressive tumor microenvironment constitutes a key barrier that greatly hinders the success of immunotherapies in the most aggressive types of breast cancer, HER2-positive and TNBC. Therefore, the improvement of the current and the demand for the development of new immunotherapeutic strategies is strongly warranted.
Collapse
|
47
|
Molecular Mechanisms of Resistance to Immunotherapy and Antiangiogenic Treatments in Clear Cell Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13235981. [PMID: 34885091 PMCID: PMC8656474 DOI: 10.3390/cancers13235981] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype arising from renal cell carcinomas. This tumor is characterized by a predominant angiogenic and immunogenic microenvironment that interplay with stromal, immune cells, and tumoral cells. Despite the obscure prognosis traditionally related to this entity, strategies including angiogenesis inhibition with tyrosine kinase inhibitors (TKIs), as well as the enhancement of the immune system with the inhibition of immune checkpoint proteins, such as PD-1/PDL-1 and CTLA-4, have revolutionized the treatment landscape. This approach has achieved a substantial improvement in life expectancy and quality of life from patients with advanced ccRCC. Unfortunately, not all patients benefit from this success as most patients will finally progress to these therapies and, even worse, approximately 5 to 30% of patients will primarily progress. In the last few years, preclinical and clinical research have been conducted to decode the biological basis underlying the resistance mechanisms regarding angiogenic and immune-based therapy. In this review, we summarize the insights of these molecular alterations to understand the resistance pathways related to the treatment with TKI and immune checkpoint inhibitors (ICIs). Moreover, we include additional information on novel approaches that are currently under research to overcome these resistance alterations in preclinical studies and early phase clinical trials.
Collapse
|
48
|
Walters AA, Dhadwar B, Al-Jamal KT. Modulating expression of inhibitory and stimulatory immune 'checkpoints' using nanoparticulate-assisted nucleic acid delivery. EBioMedicine 2021; 73:103624. [PMID: 34688033 PMCID: PMC8536530 DOI: 10.1016/j.ebiom.2021.103624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 01/15/2023] Open
Abstract
Immune checkpoints are regulatory molecules responsible for determining the magnitude and nature of the immune response. The aim of immune checkpoint targeting immunotherapy is to manipulate these interactions, engaging the immune system in treatment of cancer. Clinically, the use of monoclonal antibodies to block immunosuppressive interactions has proven itself to be a highly effective immunotherapeutic intervention. Within the literature there are numerous candidates for next generation of immune checkpoint targeting strategies. One such example is the use of nucleic acid to alter expression levels of immune checkpoint molecules, either as antisense oligo nucleotides/siRNA, to downregulate inhibitory molecules, or mRNA/DNA, to express co-stimulatory molecules. A significant component of nucleic acid delivery is its formulation within a nanoparticulate system. In this review we discuss the progress of the preclinical application of nucleic acid-based immunotherapies to target a selection of co-inhibitory/co-stimulatory molecules. Furthermore, we identify the potential and current gaps within the literature which may form the basis of future work.
Collapse
Affiliation(s)
- Adam A Walters
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Baljevan Dhadwar
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
49
|
Walters AA, Santacana-Font G, Li J, Routabi N, Qin Y, Claes N, Bals S, Tzu-Wen Wang J, Al-Jamal KT. Nanoparticle-Mediated In Situ Molecular Reprogramming of Immune Checkpoint Interactions for Cancer Immunotherapy. ACS NANO 2021; 15:17549-17564. [PMID: 34677938 PMCID: PMC8613910 DOI: 10.1021/acsnano.1c04456] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 06/01/2023]
Abstract
Immune checkpoint blockade involves targeting immune regulatory molecules with antibodies. Preclinically, complex multiantibody regimes of both inhibitory and stimulatory targets are a promising candidate for the next generation of immunotherapy. However, in this setting, the antibody platform may be limited due to excessive toxicity caused by off target effects as a result of systemic administration. RNA can be used as an alternate to antibodies as it can both downregulate immunosuppressive checkpoints (siRNA) or induce expression of immunostimulatory checkpoints (mRNA). In this study, we demonstrate that the combination of both siRNA and mRNA in a single formulation can simultaneously knockdown and induce expression of immune checkpoint targets, thereby reprogramming the tumor microenvironment from immunosuppressive to immunostimulatory phenotype. To achieve this, RNA constructs were synthesized and formulated into stable nucleic acid lipid nanoparticles (SNALPs); the SNALPs produced were 140-150 nm in size with >80% loading efficiency. SNALPs could transfect macrophages and B16F10 cells in vitro resulting in 75% knockdown of inhibitory checkpoint (PDL1) expression and simultaneously express high levels of stimulatory checkpoint (OX40L) with minimal toxicity. Intratumoral treatment with the proposed formulation resulted in statistically reduced tumor growth, a greater density of CD4+ and CD8+ infiltrates in the tumor, and immune activation within tumor-draining lymph nodes. These data suggest that a single RNA-based formulation can successfully reprogram multiple immune checkpoint interactions on a cellular level. Such a candidate may be able to replace future immune checkpoint therapeutic regimes composed of both stimulatory- and inhibitory-receptor-targeting antibodies.
Collapse
Affiliation(s)
- Adam A. Walters
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Gemma Santacana-Font
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Jin Li
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Nadia Routabi
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Yue Qin
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Nathalie Claes
- EMAT, University
of Antwerp, Groenenborgerlaan
171, 2020 Antwerp, Belgium
| | - Sara Bals
- EMAT, University
of Antwerp, Groenenborgerlaan
171, 2020 Antwerp, Belgium
| | - Julie Tzu-Wen Wang
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Khuloud T. Al-Jamal
- Institute
of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| |
Collapse
|
50
|
Bolandi N, Derakhshani A, Hemmat N, Baghbanzadeh A, Asadzadeh Z, Afrashteh Nour M, Brunetti O, Bernardini R, Silvestris N, Baradaran B. The Positive and Negative Immunoregulatory Role of B7 Family: Promising Novel Targets in Gastric Cancer Treatment. Int J Mol Sci 2021; 22:ijms221910719. [PMID: 34639059 PMCID: PMC8509619 DOI: 10.3390/ijms221910719] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer (GC), with a heterogeneous nature, is the third leading cause of death worldwide. Over the past few decades, stable reductions in the incidence of GC have been observed. However, due to the poor response to common treatments and late diagnosis, this cancer is still considered one of the lethal cancers. Emerging methods such as immunotherapy with immune checkpoint inhibitors (ICIs) have transformed the landscape of treatment for GC patients. There are presently eleven known members of the B7 family as immune checkpoint molecules: B7-1 (CD80), B7-2 (CD86), B7-H1 (PD-L1, CD274), B7-DC (PDCD1LG2, PD-L2, CD273), B7-H2 (B7RP1, ICOS-L, CD275), B7-H3 (CD276), B7-H4 (B7x, B7S1, Vtcn1), B7-H5 (VISTA, Gi24, DD1α, Dies1 SISP1), B7-H6 (NCR3LG1), B7-H7 (HHLA2), and Ig-like domain-containing receptor 2 (ILDR2). Interaction of the B7 family of immune-regulatory ligands with the corresponding receptors resulted in the induction and inhibition of T cell responses by sending co-stimulatory and co-inhibitory signals, respectively. Manipulation of the signals provided by the B7 family has significant potential in the management of GC.
Collapse
Affiliation(s)
- Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95100 Catania, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| |
Collapse
|