1
|
Fernandes J, Veldhoen M, Ferreira C. Tissue-resident memory T cells: Harnessing their properties against infection for cancer treatment. Bioessays 2024; 46:e2400119. [PMID: 39258352 DOI: 10.1002/bies.202400119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/22/2024] [Accepted: 08/26/2024] [Indexed: 09/12/2024]
Abstract
We have rapidly gained insights into the presence and function of T lymphocytes in non-lymphoid tissues, the tissue-resident memory T (TRM) cells. The central pillar of adaptive immunity has been expanded from classic central memory T cells giving rise to progeny upon reinfection and effector memory cells circulating through the blood and patrolling the tissues to include TRM cells that reside and migrate inside solid organs and tissues. Their development and maintenance have been studied in detail, providing exciting clues on how their unique properties used to fight infections may benefit therapies against solid tumors. We provide an overview of CD8 TRM cells and the properties that make them of interest for vaccination and cancer therapies.
Collapse
Affiliation(s)
- João Fernandes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Marc Veldhoen
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Cristina Ferreira
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
2
|
Li Z, He L, Li J, Qian J, Wu Z, Zhu Y, Zhuo T, Nong J, Liang H, Zheng H, Huang W, Huang J, Cao J. SASS6 promotes tumor proliferation and is associated with TP53 and immune infiltration in lung adenocarcinoma. Clin Exp Med 2024; 24:243. [PMID: 39443355 PMCID: PMC11499408 DOI: 10.1007/s10238-024-01510-0] [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: 06/15/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024]
Abstract
The most common type of non-small cell lung cancer is lung adenocarcinoma (LUAD), which is characterized by high morbidity and poor survival. Up-regulation of SASS6 expression can lead to the progression of various malignant tumors. However, there are no relevant studies on the role of SASS6 in LUAD. SASS6 was highly expressed in most tumors, reflecting a good diagnostic value, and its overexpression in LUAD indicated discouraging overall prognosis. Functional enrichment analysis suggested that SASS6 was associated with cell cycle in LUAD. In addition, patients with high SASS6 expression had worse immune infiltration, but higher TMB and immune checkpoint, and higher sensitivity to multiple targeted drugs such as osimertinib. Cell experiments confirmed that knockdown of SASS6 could inhibit the viability of tumor cells.SASS6 has important value in the diagnosis of cancer. In particular, SASS6 is a crucial factor in the progression of LUAD, and has important clinical value, especially in the diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- Zihao Li
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Lingyun He
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Jiayi Li
- Department of Nephrology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Jing Qian
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zuotao Wu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yongjie Zhu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ting Zhuo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jusen Nong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Honghua Liang
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Hua Zheng
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Weijia Huang
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China
| | - Julu Huang
- Department of Medical Oncology, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China.
| | - Jianbin Cao
- Department of Thoracic Surgery, Liuzhou People's Hospital affiliated to Guangxi Medical University, Liuzhou, Guangxi, China.
| |
Collapse
|
3
|
Golding R, Abuqubo R, Pansa CJ, Bhatta M, Shankar V, Mani K, Kleinbart E, Gelfand Y, Murthy S, De la Garza Ramos R, Krystal J, Eleswarapu A, Yassari R, Mostafa E, Fourman MS, Schlumprecht A. Immunologic and Targeted Molecular Therapies for Chordomas: A Narrative Review. J Clin Med 2024; 13:5679. [PMID: 39407739 PMCID: PMC11476405 DOI: 10.3390/jcm13195679] [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: 09/04/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
Chordomas are rare sarcomas arising from notochordal tissue and occur most commonly in the spine. The standard of care for chordomas without evidence of metastatic disease generally consists of en bloc resection followed by adjuvant radiotherapy. However, long-term (20-year) survival rates are approximately 30%. Chordomas are generally considered as chemo resistant. Therefore, systemic therapies have rarely been employed. Novel immunotherapies, including antibody therapy and tumor vaccines, have shown promise in early trials, leading to extended progression-free survival and symptom relief. However, the outcomes of larger trials using these vectors are heterogeneous. The aim of this review is to summarize novel chordoma treatments in immune-targeted therapies. The current merits, trial outcomes, and toxicities of these novel immune and targeted therapies, including those targeting vascular endothelial growth factor receptor (VEGFR) targets and the epidermal growth factor receptor (EGFR), will be discussed.
Collapse
Affiliation(s)
- Regina Golding
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| | - Rami Abuqubo
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Christopher J. Pansa
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Manish Bhatta
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Vishal Shankar
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Kyle Mani
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Emily Kleinbart
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (R.A.); (C.J.P.); (M.B.); (V.S.); (K.M.); (E.K.)
| | - Yaroslav Gelfand
- Department of Neurological Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (Y.G.); (S.M.); (R.D.l.G.R.); (R.Y.)
| | - Saikiran Murthy
- Department of Neurological Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (Y.G.); (S.M.); (R.D.l.G.R.); (R.Y.)
| | - Rafael De la Garza Ramos
- Department of Neurological Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (Y.G.); (S.M.); (R.D.l.G.R.); (R.Y.)
| | - Jonathan Krystal
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| | - Ananth Eleswarapu
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| | - Reza Yassari
- Department of Neurological Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (Y.G.); (S.M.); (R.D.l.G.R.); (R.Y.)
| | - Evan Mostafa
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| | - Mitchell S. Fourman
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| | - Anne Schlumprecht
- Department of Orthopaedic Surgery, Montefiore Einstein, Bronx, NY 10461, USA; (R.G.); (J.K.); (A.E.); (E.M.); (A.S.)
| |
Collapse
|
4
|
Su Q, Pan J, Wang C, Zhang M, Cui H, Zhao X. Curcumin and Baicalin Co-Loaded Nanoliposomes for Synergistic Treatment of Non-Small Cell Lung Cancer. Pharmaceutics 2024; 16:973. [PMID: 39204318 PMCID: PMC11359521 DOI: 10.3390/pharmaceutics16080973] [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: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 09/04/2024] Open
Abstract
Currently, the treatment of patients with advanced non-small cell lung cancer (NSCLC) mainly relies on traditional chemotherapeutic drugs; however, most of them have limited therapeutic effects and high toxicity. Some natural products with good therapeutic efficacy and low toxicity and side effects are limited in clinical application due to their low solubility and bioavailability. In this study, a nanoliposome drug-carrying system (Lip-Cur/Ba) was developed for the co-delivery of curcumin (Cur) and baicalin (Ba) using the thin-film hydration method. In vitro experiments demonstrated that Lip-Cur/Ba had a strong killing effect on A549 cells, and the inhibitory effect of Lip-Cur/Ba on A549 cells was enhanced by 67.8% and 51.9% relative to that of the single-carrier system, which could reduce the use of a single-drug dose (Lip-Cur and Lip-Ba), delay the release rate of the drug and improve the bioavailability. In vivo experiments demonstrated the antitumor activity of Lip-Cur/Ba by intravitreal injection in BALB/c mice, and there were no obvious toxic side effects. This study provides a new idea for curcumin and baicalin to be used in the co-treatment of NSCLC by constructing a new vector.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiang Zhao
- Institute of Environment and Sustainable Development in Agriculture, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.S.); (J.P.); (C.W.); (M.Z.); (H.C.)
| |
Collapse
|
5
|
Yu JJ, Everett L, Sassalos TM, Johnson MW. SEVERE MULTIFOCAL PLACOID CHORIORETINITIS ASSOCIATED WITH IMMUNE CHECKPOINT INHIBITOR THERAPY. Retin Cases Brief Rep 2024; 18:459-463. [PMID: 36913672 DOI: 10.1097/icb.0000000000001413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
PURPOSE To report a case of severe bilateral multifocal placoid chorioretinitis in a patient receiving ipilimumab and nivolumab therapy for metastatic melanoma. METHODS Retrospective, observational case report. RESULTS A 31-year-old woman on ipilimumab and nivolumab for metastatic melanoma developed severe multifocal placoid chorioretinitis in both eyes. The patient was started on topical and systemic corticosteroid therapy, and immune checkpoint inhibitor therapy was paused. After resolution of ocular inflammation, the patient was restarted on immune checkpoint inhibitor therapy without return of ocular symptoms. CONCLUSION Extensive multifocal placoid chorioretinitis may occur in patients undergoing immune checkpoint inhibitor therapy. Some patients with immune checkpoint inhibitor-related uveitis may successfully resume immune checkpoint inhibitor therapy under close collaboration with the treating oncologist.
Collapse
Affiliation(s)
- Jeffrey J Yu
- Department of Ophthalmology, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan; and
| | - Lesley Everett
- Department of Ophthalmology, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon
| | - Therese M Sassalos
- Department of Ophthalmology, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan; and
| | - Mark W Johnson
- Department of Ophthalmology, Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan; and
| |
Collapse
|
6
|
Karati D, Mukherjee S, Roy S. Deciphering the molecular mechanistic paths describing the chemotherapeutic potential and epigenetic regulation of curcumin in lung cancer: a mini review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2715-2725. [PMID: 37982888 DOI: 10.1007/s00210-023-02838-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/03/2023] [Indexed: 11/21/2023]
Abstract
In an uncontrolled inflammatory environment, the complex process of lung carcinogenesis occurs. Lung cancer remains the leading cause of cancer-related mortality worldwide. The average 5-year survival rate is still low despite significant advancements in our knowledge of lung carcinogenesis and the development of innovative therapies in recent decades. Research on adjuvant treatment, lung carcinogenesis pathways, and possible prognostic indicators has to be refocused using an innovative approach. The majority of lung cancers are discovered at an advanced stage when there is little chance of recovery. It has grown in popularity in recent years to supplement already available chemotherapeutic therapies with adjuvant herbal medications, which may lessen toxicity and adverse effects without sacrificing therapeutic efficiency. One such prospective contender is curcumin. In-depth research has been done on curcumin as a multi-target anti-tumor and anti-inflammatory molecule. A pharmacologically active polyphenol produced from turmeric is called curcumin. Over the past few decades, curcumin's therapeutic potential has been thoroughly studied, and data indicate that curcumin may play a part in a variety of biological processes, most notably its potent anticancer activity. Being a pleiotropic chemical, curcumin regulates a variety of molecules that are key players in many cell signaling pathways. It has been shown to stifle transformation, restrain proliferation, and trigger apoptosis. Curcumin can reduce the development of non-small cell LC by downregulating Circular RNA hsa_circ_0007580, which in turn controls the expression of integrin subunit beta 1 by adsorbing miR-384. Nevertheless, despite all these advantages, curcumin's effectiveness is still restricted because of its weak bioavailability, poor absorption within the systemic circulation, and quick removal from the body. In an effort to overcome these constraints, scientists from all around the world are working to develop a synthetic and improved curcuminoid by appropriately altering the parent skeleton structurally. These curcuminoids will simultaneously improve the physicochemical properties and efficacy. This review presents evidence from the most recent clinical trials coupled with the molecular mechanisms of curcumin in LC.
Collapse
Affiliation(s)
- Dipanjan Karati
- Department of Pharmaceutical Technology, School of Pharmacy, Techno India University, Kolkata, 700091, West Bengal, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata, 124 B.L. Saha Road, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
7
|
Li ZH, Li JY, Zhu YJ, Dai L, Wu ZT, Nong JS, Zhuo T, Li FL, He LY, Liang HH, Zang FL, Wang YY, Chen MW, Huang WJ, Cao JB. Analysis of Nucleoporin 107 Overexpression and Its Association with Prognosis and Immune Infiltration in Lung Adenocarcinoma by Bioinformatics Methods. Int J Gen Med 2023; 16:5449-5465. [PMID: 38021066 PMCID: PMC10676695 DOI: 10.2147/ijgm.s441185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023] Open
Abstract
Background Lung adenocarcinoma (LUAD) has high morbidity and mortality. Current studies indicate nucleoporin 107 (NUP107) is involved in the construction of nuclear pore complex, and NUP107 overexpression contributes to the growth and development in most types of cancers, but its effect in LUAD has not been elucidated. Methods Differences in NUP107 expression were investigated using the Cancer Genome Atlas (TCGA) and multiple Gene Expression Omnibus (GEO) data sets. Enrichment analysis were implemented to probe the NUP107 function. The association of NUP107 with the degree of immune cell infiltration was investigated by the TIMER database, single-sample gene set enrichment analysis (ssGSEA), and ESTIMATE. The association of NUP107 expression with tumor mutation burden (TMB), TP53, and immune checkpoint was analyzed. Single-cell RNA sequencing data were used to detect NUP107 expression in different cell clusters. Finally, we performed real-time quantitative polymerase chain reaction (RT-qPCR) and immunohistochemistry (IHC) to prove the difference of NUP107 expression. Results NUP107 was overexpressed in LUAD and mainly expressed in cancer stem cell (CSC). Overexpression of NUP107 in LUAD suggested a poorer prognosis. Functional enrichment analysis pointed out that NUP107 was mainly linked to the regulation of cell cycle. Both immune cell infiltration and TMB were found to be in connection with NUP107. Cases in the group with high NUP107 expression had poorer immune infiltration, but had higher expression of immune checkpoints, TMB, and proportion of TP53 mutations. Conclusion NUP107 is a sensitive diagnostic and prognostic factor for LUAD and may be involved in tumor progression through its effects on cell cycle and immune infiltration.
Collapse
Affiliation(s)
- Zi-Hao Li
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Jia-Yi Li
- Department of Nephrology, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Yong-Jie Zhu
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Lei Dai
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Zuo-Tao Wu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Ju-Sen Nong
- Department of Pediatric Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Ting Zhuo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Fu-Li Li
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Ling-Yun He
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Hong-Hua Liang
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Feng-Ling Zang
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Yong-Yong Wang
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Ming-Wu Chen
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, People’s Republic of China
| | - Wei-Jia Huang
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| | - Jian-Bin Cao
- Department of Thoracic Surgery, Liuzhou People’s Hospital Affiliated to Guangxi Medical University, Liuzhou, Guangxi, People’s Republic of China
| |
Collapse
|
8
|
Gajjar S, Bora V, Patel BM. Repositioning of simvastatin for diabetic colon cancer: role of CDK4 inhibition and apoptosis. Mol Cell Biochem 2023; 478:2337-2349. [PMID: 36703094 DOI: 10.1007/s11010-023-04663-w] [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: 02/14/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023]
Abstract
There is increased risk of colon cancer in both men and women having diabetes. The objective of the study was to evaluate the role of simvastatin in colon cancer associated with type 2 diabetes mellitus. Diabetes was induced by administering high fat diet with low dose streptozotocin model. 1,2 dimethylhydrazine (25 mg/kg, sc) was used for colon cancer induction. MTT assay, scratch assay, clonogenic assay and annexin V-FITC assay using flow cytometry were performed on HCT-15 cell line. Simvastatin controlled diabetes and colon cancer in animal models and reduced mRNA expression of CDK4 in colon tissues. In vitro studies revealed that simvastatin showed a decrease in cell viability and produced dose dependent decrease in clone formation. There was decrease in the rate of migration with increase in concentration of simvastatin in scratch assay. Moreover, simvastatin induced apoptosis as depicted from annexin V-FITC assay using flow cytometry as well as that revealed by tunnel assay. Our data suggest that simvastatin exhibits protective role in colon cancer associated with diabetes mellitus and acts possibly via down regulation of CDK4 and induction of apoptosis and hence can be considered for repositioning in diabetic colon cancer.
Collapse
Affiliation(s)
- Saumitra Gajjar
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | - Vivek Bora
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | | |
Collapse
|
9
|
Xu J, Shi Q, Wang B, Ji T, Guo W, Ren T, Tang X. The role of tumor immune microenvironment in chordoma: promising immunotherapy strategies. Front Immunol 2023; 14:1257254. [PMID: 37720221 PMCID: PMC10502727 DOI: 10.3389/fimmu.2023.1257254] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/14/2023] [Indexed: 09/19/2023] Open
Abstract
Chordoma is a rare malignant bone tumor with limited therapeutic options, which is resistant to conventional chemotherapy and radiotherapy, and targeted therapy is also shown with little efficacy. The long-standing delay in researching its mechanisms of occurrence and development has resulted in the dilemma of no effective treatment targets and no available drugs in clinical practice. In recent years, the role of the tumor immune microenvironment in driving tumor growth has become a hot and challenging topic in the field of cancer research. Immunotherapy has shown promising results in the treatment of various tumors. However, the study of the immune microenvironment of chordoma is still in its infancy. In this review, we aim to present a comprehensive reveal of previous exploration on the chordoma immune microenvironment and propose promising immunotherapy strategies for chordoma based on these characteristics.
Collapse
Affiliation(s)
- Jiuhui Xu
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Qianyu Shi
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Boyang Wang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tao Ji
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Tingting Ren
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| | - Xiaodong Tang
- Department of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
10
|
Peer CJ, Schmidt KT, Arisa O, Richardson WJ, Paydary K, Goldstein DA, Gulley JL, Figg WD, Ratain MJ. In Silico Re-Optimization of Atezolizumab Dosing Using Population Pharmacokinetic Simulation and Exposure-Response Simulation. J Clin Pharmacol 2023; 63:672-680. [PMID: 36624662 PMCID: PMC10175103 DOI: 10.1002/jcph.2203] [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: 10/06/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023]
Abstract
Atezolizumab, a humanized monoclonal antibody against programmed cell death ligand 1 (PD-L1), was initially approved in 2016, around the same time that the sponsor published the minimum serum concentration to maintain the saturation of receptor occupancy (6 μg/mL). The initially approved dose regimen of 1200 mg every 3 weeks (q3w) was subsequently modified to 840 mg q2w or 1680 mg q4w through pharmacokinetic simulations. Yet, each standard regimen yields steady-state trough concentrations (CMIN,SS ) far exceeding (≈ 40-fold) the stated target concentration. Additionally, the steady-state area under the plasma drug concentration-time curve (AUCSS ) at 1200 mg q3w was significantly (P = .027) correlated with the probability of adverse events of special interest (AESIs) in patients with non-small cell lung cancer (NSCLC) and, coupled with excess exposure, this provides incentive to explore alternative dose regimens to lower the exposure burden while maintaining an effective CMIN,SS . In this study, we first identified 840 mg q6w as an extended-interval regimen that could robustly maintain a serum concentration of 6 μg/mL (≥99% of virtual patients simulated, n = 1000), then applied this regimen to an approach that administers 2 "loading doses" of standard-interval regimens for a future clinical trial aiming to personalize dose regimens. Each standard dose was simulated for 2 loading doses, then 840 mg q6w thereafter; all yielded cycle-7 CMIN,SS values of >6 μg/mL in >99% of virtual patients. Further, the AUCSS from 840 mg q6w resulted in a flattening (P = .63) of the exposure-response relationship with adverse events of special interest (AESIs). We next aim to verify this in a clinical trial seeking to validate extended-interval dosing in a personalized approach using therapeutic drug monitoring.
Collapse
Affiliation(s)
- Cody J. Peer
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Keith T. Schmidt
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Oluwatobi Arisa
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | | | - Koosha Paydary
- Department of Medicine, Section of Hematology/Oncology, and Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| | - Daniel A. Goldstein
- Faculty of Medicine, Tel Aviv University, Israel
- Davidoff Cancer Center, Rabin Medical Center, Israel
- Clalit Health Services, Tela Aviv, Israel
| | - James L. Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, Bethesda, MD, USA
| | - William D. Figg
- Clinical Pharmacology Program, National Cancer Institute, Bethesda, MD, USA
| | - Mark J. Ratain
- Department of Medicine, Section of Hematology/Oncology, and Committee on Clinical Pharmacology & Pharmacogenomics, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Qi P, Qi B, Ding Y, Sun J, Gu C, Huo S, Liu Y, Zhao B. Implications of obstructive sleep apnea in lung adenocarcinoma: A valuable omission in cancer prognosis and immunotherapy. Sleep Med 2023; 107:268-280. [PMID: 37263079 DOI: 10.1016/j.sleep.2023.05.013] [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: 01/13/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/03/2023]
Abstract
Lung adenocarcinoma (LUAD) is a highly invasive malignant tumor with poor prognosis, and there is growing evidence that obstructive sleep apnea (OSA) could significantly promotes the risk of LUAD. In order to improve the treatment outcomes of patients with LUAD and OSA, we aim to screen OSA-related genes that may potentially affect LUAD and to discover a high sensitivity prognostic signature that can stratify LUAD/OSA patients and to further accurately identify LUAD patients who might respond to immunotherapy. Molecular subtypes classified by the prognostic signature did not belong to any previously reported subtypes of LUAD. The tumor microenvironment (TME), mutation, and so on, were significantly distinct between patients within different risk groups or clusters. Combined with gene set variation analysis (GSVA) and drug susceptibility analysis, patients in the low-risk group (The vast majority of patients belonging to cluster2 by molecular subtyping) were not suitable for immunotherapy due to T-cell exhaustion caused by long-term inflammatory response; the question of how to reverse T-cell exhaustion may be a primary consideration. Cluster3 patients had the highest benefit from immunotherapy, and although cluster1 patients had the worst prognosis, they were more sensitive to traditional chemotherapeutic drugs. Animal experiments showed that chronic intermittent hypoxia (CIH) could not only significantly promote the tumor growth of LUAD, but also increase the expression levels of risk genes. This risk model may contribute greatly to the evaluation of prognosis, molecular characteristics, and treatment modalities of LUAD/OSA, and could be further translated into clinical applications to ameliorate the treatment dilemmas.
Collapse
Affiliation(s)
- Pengju Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Bo Qi
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Yuan Ding
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Jianxia Sun
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Chengwei Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Shuhua Huo
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Yuzhen Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China; Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China.
| | - Baosheng Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China; Esophageal Cancer Institute of Xinxiang Medical University, Weihui, 453100, Henan, China.
| |
Collapse
|
12
|
Mishra AP, Singh P, Yadav S, Nigam M, Seidel V, Rodrigues CF. Role of the Dietary Phytochemical Curcumin in Targeting Cancer Cell Signalling Pathways. PLANTS (BASEL, SWITZERLAND) 2023; 12:plants12091782. [PMID: 37176840 PMCID: PMC10180989 DOI: 10.3390/plants12091782] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/19/2023] [Accepted: 04/22/2023] [Indexed: 05/15/2023]
Abstract
The diarylheptanoid curcumin [(1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione] is one of the phenolic pigments responsible for the yellow colour of turmeric (Curcuma longa L.). This phytochemical has gained much attention in recent years due to its therapeutic potential in cancer. A range of drug delivery approaches have been developed to optimise the pharmacokinetic profile of curcumin and ensure that it reaches its target sites. Curcumin exhibits numerous biological effects, including anti-inflammatory, cardioprotective, antidiabetic, and anti-aging activities. It has also been extensively studied for its role as a cancer chemopreventive and anticancer agent. This review focusses on the role of curcumin in targeting the cell signalling pathways involved in cancer, particularly via modulation of growth factors, transcription factors, kinases and other enzymes, pro-inflammatory cytokines, and pro-apoptotic and anti-apoptotic proteins. It is hoped that this study will help future work on the potential of curcumin to fight cancer.
Collapse
Affiliation(s)
- Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, Bloemfontein 9300, South Africa
| | - Pratichi Singh
- Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Shikha Yadav
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida 203201, Uttar Pradesh, India
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal University, Srinagar Garhwal 246174, Uttarakhand, India
| | - Veronique Seidel
- Natural Products Research Laboratory, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK
| | - Celia Fortuna Rodrigues
- LEPABE-Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE-Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- TOXRUN-Toxicology Research Unit, Cooperativa de Ensino Superior Politécnico e Universitário-CESPU, 4585-116 Gandra PRD, Portugal
| |
Collapse
|
13
|
Pan J, Huang T, Deng Z, Zou C. Roles and therapeutic implications of m6A modification in cancer immunotherapy. Front Immunol 2023; 14:1132601. [PMID: 36960074 PMCID: PMC10028070 DOI: 10.3389/fimmu.2023.1132601] [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: 12/27/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
Recent studies have demonstrated that N6-methyladenosine (m6A), the most abundant, dynamic, and reversible epigenetic RNA modification in eukaryotes, is regulated by a series of enzymes, including methyltransferases (writers), demethylases (erasers), and m6A recognition proteins (readers). Aberrant regulation of m6A modification is pivotal for tumorigenesis, progression, invasion, metastasis, and apoptosis of malignant tumors. Immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, as recognized by the 2018 Nobel Prize in Medicine and Physiology. However, not all cancer patients response to ICI therapy, which is thought to be the result of intricate immune escape mechanisms. Recently, numerous studies have suggested a novel role for m6A epigenetic modification in the regulation of tumor immune evasion. Herein, we review the relevant mechanisms of m6A regulators in regulating various key signaling pathways in cancer biology and how m6A epigenetic modifications regulate the expression of immune checkpoints, opening a new window to understand the roles and mechanisms of m6A epigenetic modifications in regulating tumor immune evasion. In addition, we highlight the prospects and development directions of future combined immunotherapy strategies based on m6A modification targeting, providing directions for promoting the treatment outcomes of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Juan Pan
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
- Department of Clinical Medical Research Center, The 2nd Clinical Medical College (Shenzhen People’s Hospital) of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Tuxiong Huang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhenjun Deng
- Department of Dermatology, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), Shenzhen, China
- The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Chang Zou
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, China
- Department of Clinical Medical Research Center, The 2nd Clinical Medical College (Shenzhen People’s Hospital) of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Shenzhen Public Service Platform On Tumor Precision Medicine and Molecular Diagnosis, The Second Clinical Medical College of Jinan University, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
14
|
A systematic review and meta-analysis of immune checkpoint therapy in relapsed or refractory non-Hodgkin lymphoma; a friend or foe? Transl Oncol 2023; 30:101636. [PMID: 36773442 PMCID: PMC9941575 DOI: 10.1016/j.tranon.2023.101636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 01/11/2023] [Accepted: 01/30/2023] [Indexed: 02/11/2023] Open
Abstract
Over the last decades, a revolution has occurred in oncology with the development of immune checkpoint inhibitors (ICIs). Following tremendous successes in solid tumors, interest has risen to explore these inhibitors in hematologic malignancies; while Hodgkin's lymphoma (HL) has shown overwhelming achievements, available data on different types of non-Hodgkin's lymphoma (NHL) vary considerably. To the best of our knowledge, no meta-analysis has assessed the efficacy and safety of ICI therapy in relapsed or refractory NHL patients. Meta-analysis of the included studies (n = 29) indicated PD-1 may probably be the more attractive ICI target rather than PD-L1 and CTLA-4 in NHL patients. Also, there is a plausible correlation between NHL subtypes and response to ICI therapy. While MF, ENKTL, RT, and PMBCL showed promising responses to ICI monotherapy, neither FL nor DLBCL had satisfactory responses; further necessitating novel strategies such as the application of ICIs in combination with other treatment strategies. Notably, among different combinations, BTK inhibitors showed an obvious improvement as compared to ICI monotherapy in both FL and DLBCL, however, the best results were obtained when ICI was combined with anti-CD20 monoclonal antibodies. Finally, while most NHL patients who received ICI treatment have experienced mild AEs, larger trials with long-term follow-up are required to confirm the safety, as well as the efficacy, of ICI therapy in NHL patients.
Collapse
|
15
|
Li Z, Zheng Y, Wu Z, Zhuo T, Zhu Y, Dai L, Wang Y, Chen M. NCAPD2 is a novel marker for the poor prognosis of lung adenocarcinoma and is associated with immune infiltration and tumor mutational burden. Medicine (Baltimore) 2023; 102:e32686. [PMID: 36701707 PMCID: PMC9857258 DOI: 10.1097/md.0000000000032686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Lung adenocarcinoma (LUAD) is at present the most prevalent subtype of lung cancer worldwide. Non-SMC condensin I complex subunit D2 (NCAPD2) is one of the 3 non-SMC subunits in condensin I. Previous studies have confirmed that NCAPD2 plays a critical role in chromosome cohesion and segregation. NCAPD2 may be involved in tumorigenesis and progression by participating in abnormal cell cycle division, but the prognostic value of NCAPD2 in LUAD remains unclear. We investigated differences in the expression levels of NCAPD2 and determined their association with clinical features, as well as their diagnostic and prognostic value using the cancer genome atlas database. The function of NCAPD2 was analyzed using gene ontology, Kyoto encyclopedia of genes and genomes, and gene set enrichment analysis. CIBERSORT, single-sample gene set enrichment analysis, and ESTIMATE were used to analyze the immune microenvironment of tumor patients. Tumor mutational burden (TMB) and immune checkpoints were analyzed, while hub genes were identified using weighted gene coexpression network analysis and were used to construct prognostic models. Subsequently, the competing endogenous RNAs network of NCAPD2 in LUAD was explored. Finally, we performed qPCR to verify differences in NCAPD2 expression between the tumor and normal tissues. The expression of NCAPD2 in LUAD was significantly upregulated compared with normal lung tissues. NCAPD2 has been linked to the T stage, N stage, and tumor stage. The elevated expression of NCAPD2 in LUAD can predict a poor prognosis. Functional enrichment analysis indicated that the main function of NCAPD2 was in cell cycle regulation. Moreover, NCAPD2 was also associated with immune cell infiltration and TMB. NCAPD2 is a novel prognostic marker in LUAD and is associated with immune infiltration and TMB.
Collapse
Affiliation(s)
- Zihao Li
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yuxuan Zheng
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zuotao Wu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ting Zhuo
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yongjie Zhu
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lei Dai
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yongyong Wang
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mingwu Chen
- Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- * Correspondence: Mingwu Chen, Department of Cardio-Thoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China (e-mail: )
| |
Collapse
|
16
|
Rekulapelli A, E. Flausino L, Iyer G, Balkrishnan R. Effectiveness of immunological agents in non-small cell lung cancer. Cancer Rep (Hoboken) 2022; 6:e1739. [PMID: 36289059 PMCID: PMC9981233 DOI: 10.1002/cnr2.1739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 08/28/2022] [Accepted: 10/08/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND AND AIM Non-small cell lung cancer (NSCLC) continues to claim millions of lives worldwide. Although its poor prognosis is largely attributed to the lack of adequate and precise detection technologies, cancer cells' suppression of the immune system adds on to the difficulty of identifying abnormal NSCLC tumors in their early stages. Therefore, cancer immunotherapy, which activates the immune system and helps it fight tumors, has recently become the most sought-after technique, especially in the advanced stages of NSCLC, where surgery or chemotherapy may or may not bring about the desired survival benefits in patients. METHODS This review focuses on the various immunotherapeutic interventions and their efficacy in advanced NSCLC clinical trials. Monoclonal antibodies like anti-PD-1/PD-L1 agents and anti-CTLA-4 antibodies, cancer vaccines, oncolytic viruses and adoptive T cell therapy have been discussed in brief. Furthermore, the effects of gender, age, and race on the efficacy of immune checkpoint inhibitors and suggest plausible future approaches in the realm of immuno-oncology. RESULTS Immunotherapy is used alone or in combination either with other immunological agents or with chemotherapy. However, the efficacy of these strategies depends extensively on various demographic variables, as some patients respond perfectly well to immunotherapy, while others do not benefit at all or experience disease progression. By targeting a "hallmark" of cancer (immune evasion), immunotherapy has transformed NSCLC management, though several barriers prevent its complete effectiveness. CONCLUSIONS All these immunological strategies should be interpreted in the current setting of synergistic treatment, in which these agents can be combined with chemotherapy, radiotherapy, and, or surgery following patient and tumor characteristics to proportionate the best-individualized treatment and achieve superior results. To better pursue this goal, further investigations on cost-effectiveness and sex-gender, race, and age differences in immunotherapy are needed.
Collapse
Affiliation(s)
- Akhil Rekulapelli
- Department of Public Health SciencesUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| | - Lucas E. Flausino
- Department of Public Health SciencesUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA,Faculdade de MedicinaUniversidade de São PauloSão PauloBrazil
| | - Gayatri Iyer
- Department of Pharmaceutical Sciences and TechnologyInstitute of Chemical TechnologyMumbaiIndia
| | - Rajesh Balkrishnan
- Department of Public Health SciencesUniversity of Virginia School of MedicineCharlottesvilleVirginiaUSA
| |
Collapse
|
17
|
Shah D, Joshi M, Patel BM. Role of NIMA‐related kinase 2 in lung cancer: Mechanisms and therapeutic prospects. Fundam Clin Pharmacol 2022; 36:766-776. [DOI: 10.1111/fcp.12777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/10/2022] [Accepted: 03/24/2022] [Indexed: 01/04/2023]
Affiliation(s)
- Darshak Shah
- Institute of Pharmacy Nirma University Ahmedabad India
| | - Mit Joshi
- Institute of Pharmacy Nirma University Ahmedabad India
| | | |
Collapse
|
18
|
Hu Y, Paris S, Bertolet G, Barsoumian HB, He K, Sezen D, Chen D, Wasley M, Silva JDA, Mitchell JA, Voss TA, Masrorpour F, Leyton CK, Yang L, Leuschner C, Puebla-Osorio N, Gandhi S, Nguyen QN, Cortez MA, Welsh JW. Combining a nanoparticle-mediated immunoradiotherapy with dual blockade of LAG3 and TIGIT improves the treatment efficacy in anti-PD1 resistant lung cancer. J Nanobiotechnology 2022; 20:417. [PMID: 36123677 PMCID: PMC9484155 DOI: 10.1186/s12951-022-01621-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 08/26/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND While improvements in immunoradiotherapy have significantly improved outcomes for cancer patients, this treatment approach has nevertheless proven ineffective at controlling the majority of malignancies. One of the mechanisms of resistance to immunoradiotherapy is that immune cells may be suppressed via the myriad of different immune checkpoint receptors. Therefore, simultaneous blockade of multiple immune checkpoint receptors may enhance the treatment efficacy of immunoradiotherapy. METHODS We combined NBTXR3-enhanced localized radiation with the simultaneous blockade of three different checkpoint receptors: PD1, LAG3, and TIGIT, and tested the treatment efficacy in an anti-PD1-resistant lung cancer model in mice. 129 Sv/Ev mice were inoculated with fifty thousand αPD1-resistant 344SQR cells in the right leg on day 0 to establish primary tumors and with the same number of cells in the left leg on day 4 to establish the secondary tumors. NBTXR3 was intratumorally injected into the primary tumors on day 7, which were irradiated with 12 Gy on days 8, 9, and 10. Anti-PD1 (200 µg), αLAG3 (200 µg), and αTIGIT (200 µg) were given to mice by intraperitoneal injections on days 5, 8, 11, 14, 21, 28, 35, and 42. RESULTS This nanoparticle-mediated combination therapy is effective at controlling the growth of irradiated and distant unirradiated tumors, enhancing animal survival, and is the only one that led to the destruction of both tumors in approximately 30% of the treated mice. Corresponding with this improved response is robust activation of the immune response, as manifested by increased numbers of immune cells along with a transcriptional signature of both innate and adaptive immunity within the tumor. Furthermore, mice treated with this combinatorial therapy display immunological memory response when rechallenged by the same cancer cells, preventing tumor engraftment. CONCLUSION Our results strongly attest to the efficacy and validity of combining nanoparticle-enhanced radiotherapy and simultaneous blockade of multiple immune checkpoint receptors and provide a pre-clinical rationale for investigating its translation into human patients.
Collapse
Affiliation(s)
- Yun Hu
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Sébastien Paris
- Department of Translational Science, Nanobiotix, Paris, France
| | - Genevieve Bertolet
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Hampartsoum B Barsoumian
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Kewen He
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Duygu Sezen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Dawei Chen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Mark Wasley
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Jordan DA Silva
- Department of Translational Science, Nanobiotix, Paris, France
| | - Joylise A Mitchell
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Tiffany A Voss
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Fatemeh Masrorpour
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Claudia Kettlun Leyton
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Liangpeng Yang
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Carola Leuschner
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Nahum Puebla-Osorio
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Saumil Gandhi
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Quynh-Nhu Nguyen
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - Maria Angelica Cortez
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| | - James W Welsh
- Department of Radiation Oncology, Unit 97, The University of Texas MD Anderson Cancer, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Kuo AM, Kraehenbuehl L, King S, Leung DYM, Goleva E, Moy AP, Lacouture ME, Shah NJ, Faleck DM. Contribution of the Skin-Gut Axis to Immune-Related Adverse Events with Multi-System Involvement. Cancers (Basel) 2022; 14:cancers14122995. [PMID: 35740660 PMCID: PMC9221505 DOI: 10.3390/cancers14122995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/12/2022] [Accepted: 06/12/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Increasing numbers of cancer patients are treated with immunotherapy that activates their immune systems to control or even eliminate tumors. However, a substantial proportion of patients experience adverse events mediated by the unleashed immune system. The skin is one of the most frequently affected organs, with toxicities typically manifesting as distinct types of rashes. The gastrointestinal (GI) tract is also commonly affected, with a wide spectrum of symptom manifestations that can range from self-limited diarrhea to life-threatening colitis. Here we present the relationship between skin and GI adverse events among cancer patients receiving treatment with immune checkpoint blockade, which has not been well-studied. Abstract Immune-related adverse events (irAEs) frequently complicate treatment with immune checkpoint blockade (ICB) targeting CTLA-4, PD-1, and PD-L1, which are commonly used to treat solid and hematologic malignancies. The skin and gastrointestinal (GI) tract are most frequently affected by irAEs. While extensive efforts to further characterize organ-specific adverse events have contributed to the understanding and management of individual toxicities, investigations into the relationship between multi-organ toxicities have been limited. Therefore, we aimed to conduct a characterization of irAEs occurring in both the skin and gut. A retrospective analysis of two cohorts of patients treated with ICB at Memorial Sloan Kettering Cancer Center was conducted, including a cohort of patients with cutaneous irAEs (ircAEs) confirmed by dermatologists (n = 152) and a cohort of patients with biopsy-proven immune-related colitis (n = 246). Among both cohorts, 15% (61/398) of patients developed both skin and GI irAEs, of which 72% (44/61) patients had ircAEs preceding GI irAEs (p = 0.00013). Our study suggests that in the subset of patients who develop both ircAEs and GI irAEs, ircAEs are likely to occur first. Further prospective studies with larger sample sizes are needed to validate our findings, to assess the overall incidence of co-incident irAEs, and to determine whether ircAEs are predictors of other irAEs. This analysis highlights the development of multi-system dermatologic and gastrointestinal irAEs and underscores the importance of oncologists, gastroenterologists, and dermatologists confronted with an ircAE to remain alert for additional irAEs.
Collapse
Affiliation(s)
- Alyce M. Kuo
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (A.M.K.); (M.E.L.)
| | - Lukas Kraehenbuehl
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (A.M.K.); (M.E.L.)
- Ludwig Collaborative and Swim Across America Laboratory, Parker Institute for Cancer Immunotherapy, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence: or
| | - Stephanie King
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (S.K.); (D.M.F.)
| | - Donald Y. M. Leung
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health Hospital, Denver, CO 80206, USA; (D.Y.M.L.); (E.G.)
| | - Elena Goleva
- Division of Allergy-Immunology, Department of Pediatrics, National Jewish Health Hospital, Denver, CO 80206, USA; (D.Y.M.L.); (E.G.)
| | - Andrea P. Moy
- Dermatopathology Service, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Mario E. Lacouture
- Dermatology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; (A.M.K.); (M.E.L.)
| | - Neil J. Shah
- Genitourinary Solid Tumor Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - David M. Faleck
- Gastroenterology, Hepatology & Nutrition Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (S.K.); (D.M.F.)
| |
Collapse
|
20
|
Wang Y, Duan M, Peng Z, Fan R, He Y, Zhang H, Xiong W, Jiang W. Advances of DNA Damage Repair-Related Drugs and Combination With Immunotherapy in Tumor Treatment. Front Immunol 2022; 13:854730. [PMID: 35281059 PMCID: PMC8904426 DOI: 10.3389/fimmu.2022.854730] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 01/31/2022] [Indexed: 12/21/2022] Open
Abstract
Cancer therapy has been an important and popular area in cancer research. With medical technology developing, the appearance of various targeted drugs and immunotherapy offer more choices to cancer treatment. With the increase in drug use, people have found more and more cases in which tumors are resistant to DNA damage repair (DDR)-based drugs. Recently, the concept of combination therapy has been brought up in cancer research. It takes advantages of combining two or more therapies with different mechanisms, aiming to benefit from the synergistic effects and finally rescue patients irresponsive to single therapies. Combination therapy has the potential to improve current treatment of refractory and drug-resistant tumors. Among the methods used in combination therapy, DDR is one of the most popular methods. Recent studies have shown that combined application of DDR-related drugs and immunotherapies significantly improve the therapeutic outcomes of malignant tumors, especially solid tumors.
Collapse
Affiliation(s)
- Yumin Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Meihan Duan
- School of Medicine, Tsinghua University, Beijing, China
| | - Zhouying Peng
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Ruohao Fan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yuxiang He
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China.,Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Hua Zhang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiong
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Weihong Jiang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| |
Collapse
|
21
|
Cardiac Complications: The Understudied Aspect of Cancer Cachexia. Cardiovasc Toxicol 2022; 22:254-267. [PMID: 35171467 DOI: 10.1007/s12012-022-09727-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022]
Abstract
The global burden of cancer cachexia is increasing along with drastic increase in cancer patients. Cancer itself leads to cachexia, and cachexia development is associated with events like altered hemodynamics, and reduced functional capacity of the heart among others which lead to failure of the heart and are called cardiovascular complications associated with cancer cachexia. In some patients, the anti-cancer therapy also leads to this cardiovascular complications. So, in this review, an attempt is made to understand the mechanisms, pathophysiology of cardiovascular events in cachectic patients. Important processes which cause cardiovascular complications include alterations in the structure of the heart, loss of cardiac mass and functioning, cardiac fibrosis and cardiac remodeling, apoptosis, cardiac muscle atrophy, and mitochondrial alterations. Previously, the available treatment options were limited to nutraceuticals and physical exercise. Recently, studies with some prospective agents that can improve cardiac health have been reported, but whether their action is effective in cardiovascular complications associated with cancer cachexia is not known or are under trial.
Collapse
|
22
|
Zheng Y, Han L, Chen Z, Li Y, Zhou B, Hu R, Chen S, Xiao H, Ma Y, Xie G, Yang J, Ding X, Shen L. PD-L1+CD8+ T cells enrichment in lung cancer exerted regulatory function and tumor-promoting tolerance. iScience 2022; 25:103785. [PMID: 35146396 PMCID: PMC8819393 DOI: 10.1016/j.isci.2022.103785] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 12/09/2021] [Accepted: 01/12/2022] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy targeting checkpoint blockade to rescue T cells from exhaustion has become an essential therapeutic strategy in treating cancers. Till now, little is known about the PD-L1 graphic pattern and characteristics in CD8+ T cells. We combined cytometry by time-of-flight (CyTOF) and imaging mass cytometry (IMC) approaches to analyze CD8+ T cells from primary lung cancers and discovered that PD-L1+CD8+ T cells were enriched in tumor lesions, spatially localized with PD-1+CD8+ T cells. Furthermore, PD-L1+CD8+ T cells exerted regulatory functions that inhibited CD8+ T cells proliferation and cytotoxic abilities through the PD-L1/PD-1 axis. Moreover, tumor-derived IL-27 promotes PD-L1+CD8+ T cells development through STAT1/STAT3 signaling. Single-cell RNA sequencing data analysis further clarified PD-L1+CD8+ T cells elevated in the components related to downregulation of adaptive immune response. Collectively, our data demonstrated that PD-L1+CD8+ T cells enriched in lung cancer engaged in tolerogenic effects and may become a therapeutic target in lung cancer. CyTOF and IMC revealed PD-L1+CD8+ T cells were enriched in human lung cancer PD-L1+CD8+ T cells inhibited CD8+ T cells function through PD-1/PD-L1 axis IL27 promoted PD-L1+CD8 T cells development through STAT1/STAT3 signaling
Collapse
Affiliation(s)
- Yingxia Zheng
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Corresponding author
| | - Li Han
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Zheyi Chen
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yiyang Li
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Bingqian Zhou
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Rui Hu
- Department of Thoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200230, China
| | - Shiyu Chen
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Haibo Xiao
- Department of Thoracic Surgery, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200230, China
| | - Yanhui Ma
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Guohua Xie
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Junyao Yang
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Xianting Ding
- State Key Laboratory of Oncogenes and Related Genes, School of Biomedical Engineering, Institute for Personalized Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
- Corresponding author
| | - Lisong Shen
- Department of Laboratory Medicine, Xin Hua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Faculty of Medical Laboratory Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Xin Hua Children’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
- Corresponding author
| |
Collapse
|
23
|
Cui M, Xia Q, Zhang X, Yan W, Meng D, Xie S, Shen S, Jin H, Wang S. Development and Validation of a Tumor Mutation Burden-Related Immune Prognostic Signature for Ovarian Cancers. Front Genet 2022; 12:688207. [PMID: 35087563 PMCID: PMC8787320 DOI: 10.3389/fgene.2021.688207] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 12/22/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer (OC), one of the most common malignancies of the female reproductive system, is characterized by high incidence and poor prognosis. Tumor mutation burden (TMB), as an important biomarker that can represent the degree of tumor mutation, is emerging as a key indicator for predicting the efficacy of tumor immunotherapy. In our study, the gene expression profiles of OC were downloaded from TCGA and GEO databases. Subsequently, we analyzed the prognostic value of TMB in OC and found that a higher TMB score was significantly associated with a better prognosis (p = 0.004). According to the median score of TMB, 9 key TMB related immune prognostic genes were selected by LASSO regression for constructing a TMB associated immune risk score (TMB-IRS) signature, which can effectively predict the prognosis of OC patients (HR = 2.32, 95% CI = 1.68–3.32; AUC = 0.754). Interestingly, TMB-IRS is also closely related to the level of immune cell infiltration and immune checkpoint molecules (PD1, PD-L1, CTLA4, PD-L2) in OC. Furthermore, the nomogram combined with TMB-IRS and a variety of clinicopathological features can more comprehensively evaluate the prognosis of patients. In conclusion, we explored the relationship between TMB and prognosis and validated the TMB-IRS signature based on TMB score in an independent database (HR = 1.60, 95% CI = 1.13–2.27; AUC = 0.639), which may serve as a novel biomarker for predicting OC prognosis as well as possible therapeutic targets.
Collapse
Affiliation(s)
- Mengjing Cui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Qianqian Xia
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xing Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Wenjing Yan
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Dan Meng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Shuqian Xie
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Siyuan Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Hua Jin
- Clinical Laboratory, Affiliated Tumor Hospital of Nantong University (Nantong Tumor Hospital), Nantong, China
| | - Shizhi Wang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
24
|
Luo W, Wang Z, Zhang T, Yang L, Xian J, Li Y, Li W. Immunotherapy in non-small cell lung cancer: rationale, recent advances and future perspectives. PRECISION CLINICAL MEDICINE 2021; 4:258-270. [PMID: 35692863 PMCID: PMC8982543 DOI: 10.1093/pcmedi/pbab027] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 02/05/2023] Open
Abstract
Lung cancer, with non-small cell lung cancer (NSCLC) being the major type, is the second most common malignancy and the leading cause of cancer-related death globally. Immunotherapy, represented by immune checkpoint inhibitors (ICIs), has been one of the greatest advances in recent years for the treatment of solid tumors including NSCLC. However, not all NSCLC patients experience an effective response to immunotherapy with the established selection criteria of programmed death ligand 1 (PD-L1) and tumor mutational burden (TMB). Furthermore, a considerable proportion of patients experience unconventional responses, including pseudoprogression or hyperprogressive disease (HPD), immune-related toxicities, and primary or acquired resistance during the immunotherapy process. To better understand the immune response in NSCLC and provide reference for clinical decision-making, we herein review the rationale and recent advances in using immunotherapy to treat NSCLC. Moreover, we discuss the current challenges and future strategies of this approach to improve its efficacy and safety in treating NSCLC.
Collapse
Affiliation(s)
- Wenxin Luo
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhoufeng Wang
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Zhang
- Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Lan Yang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinghong Xian
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yalun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
- Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
25
|
Sun Y, Duan J, Fang W, Wang Z, Du X, Wang X, Li C, Cai S, Zhao J, Li S, Zhang L, Bai H, Wang J. Identification and validation of tissue or ctDNA PTPRD phosphatase domain deleterious mutations as prognostic and predictive biomarkers for immune checkpoint inhibitors in non-squamous NSCLC. BMC Med 2021; 19:239. [PMID: 34615542 PMCID: PMC8496052 DOI: 10.1186/s12916-021-02075-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/27/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND With the revolutionary progress of immune checkpoint inhibitors (ICIs) achieved in non-small cell lung cancers (NSCLC), identifying patients benefiting from ICIs becomes critical and urgent. The associations of genomic alterations in protein tyrosine phosphatase receptor-type (PTPRs) and ICIs responses are unknown. METHODS Whole-exome sequencing (WES) of 73 advanced NSCLC tumors sampled before anti-PD-(L)1 therapy was carried out with corresponding clinical data collected as a discovery cohort to find the associations of PTPR mutations and ICI responses. Three validation cohorts consolidated by 7 public cohorts of 1920 NSCLC patients with WES or target sequencing data of tumor tissue-derived DNA or circulating tumor DNA (ctDNA) and relevant clinical data were applied as validation cohorts. The lung adenocarcinoma (LUAD) cohort (n=586) in The Cancer Genome Atlas (TCGA) database was used for analyzing the potential anti-tumor immunologic mechanisms. RESULTS With the highest mutation frequency among all PTPRs, PTPRD mutations in non-squamous NSCLC (ns-NSCLC) were linked to longer progression-free survivals (PFS, 324 vs 63 days, hazard ratio (HR)=0.36, p= 0.0152) and higher objective response rate (ORR, p=0.0099). In validation cohort 1 (n=377), ns-NSCLC patients with tissue PTPRD mutations had favorable PFS (9.10 vs 4.33 months, HR=0.62, p=0.0184) and ORR (p=0.013). In validation cohort 2 (n=406), ns-NSCLC patients with tissue PTPRD mutations had favorable overall survivals (OS, over 40 vs 11.94 months, HR=0.57, p=0.011). In validation cohort 3 (n=1137), ns-NSCLC patients with ctDNA PTPRD mutations had longer PFS (6.97 vs 2.73 months, HR=0.63, p=0.028) and higher ORR (p=0.047). Moreover, it was deleterious mutations in phosphatase domains (phosphatase-mut), rather than other mutations (other-mut), that were responsible of PTPRD's prediction efficiency. In addition, in validation cohort 3, ctDNA phosphatase-mut also functioned as a predictive biomarker helping identify patients benefiting more from ICIs than chemotherapy (interaction P for PFS=0.0506, for OS=0.04). Univariate and multivariate regression analysis revealed that phosphatase-mut was independent on PD-L1 expression and tumor mutation burden (TMB) to predict. In silico analysis based on TCGA LUAD cohort discovered enhanced anti-tumor immunity in phosphatase-mut patients. CONCLUSIONS Tissue or ctDNA PTPRD phosphatase domain deleterious mutations might function as a both prognostic and predictive biomarker predicting clinical outcomes of ICIs in ns-NSCLC patients, independent on TMB or PD-L1 expression.
Collapse
Affiliation(s)
- Yiting Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Wenfeng Fang
- Medical Oncology, Sun Yat-sen University Cancer Center, 651# East Dong Feng Road, Guangzhou, 510060, Guangdong, China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Xinyang Du
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Xin Wang
- Thoracic Surgery Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengcheng Li
- The Medical Department, Burning Rock Biotech, Guangzhou, China
| | - Shangli Cai
- The Medical Department, Burning Rock Biotech, Guangzhou, China
| | - Jie Zhao
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Sini Li
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China
| | - Li Zhang
- Medical Oncology, Sun Yat-sen University Cancer Center, 651# East Dong Feng Road, Guangzhou, 510060, Guangdong, China.
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China.
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 17 Pan-jia-yuan South Lane, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
26
|
Ma LR, Li JX, Tang L, Li RZ, Yang JS, Sun A, Leung ELH, Yan PY. Immune checkpoints and immunotherapy in non-small cell lung cancer: Novel study progression, challenges and solutions. Oncol Lett 2021; 22:787. [PMID: 34594428 PMCID: PMC8456509 DOI: 10.3892/ol.2021.13048] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Lung cancer is the most common type of cancer with the highest mortality rate worldwide. Non-small cell lung cancer (NSCLC) accounts for ~85% of the total number of lung cancer cases. In the past two decades, immunotherapy has become a more promising treatment method than traditional treatments (surgery, radiotherapy and chemotherapy). Immunotherapy has been shown to improve the survival rate of patients and to have a superior effect when controlling lung cancer than traditional therapy. However, only a small number of patients can benefit from immunotherapy, and not all patients who qualify experience long-term benefits. In the clinic, the objective response rate of programmed cell death protein 1 treatment without the prior screening of patients is only 15-20%. Immunotherapy is associated with both opportunities and challenges for patients with NSCLC. The current challenges of immunotherapy include the lack of accurate biomarkers, inevitable resistance and insufficient understanding of immune checkpoints. In previous years, several methods for overcoming the challenges posed by immunotherapy have been proposed, but combination therapy is the most suitable choice. A large number of studies have shown that the combination of drugs can significantly improve their efficacy, compared with monotherapy, and that some therapeutic combinations have been approved by the Food and Drug Administration for the treatment of NSCLC. Traditional Chinese medicine (TCM) is a traditional medical practice in China that can play an important role in immunotherapy. Most agents used in TCM originate from plants, and have the advantages of low toxicity and multiple targets. In addition, TCM includes a unique class of drugs that can improve autoimmunity. Therefore, TCM may be a promising treatment method for all types of cancer.
Collapse
Affiliation(s)
- Lin-Rui Ma
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Jia-Xin Li
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Ling Tang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China.,School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Run-Ze Li
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Jia-Shun Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510630, P.R. China
| | - Ao Sun
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| | - Elaine Lai-Han Leung
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China.,Department of Integrated Chinese and Western Medicine, Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai 519000, P.R. China
| | - Pei-Yu Yan
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau 999078, P.R. China
| |
Collapse
|
27
|
Dhar R, Seethy A, Singh S, Pethusamy K, Srivastava T, Talukdar J, Rath GK, Karmakar S. Cancer immunotherapy: Recent advances and challenges. J Cancer Res Ther 2021; 17:834-844. [PMID: 34528529 DOI: 10.4103/jcrt.jcrt_1241_20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Immunotherapy is a treatment that uses specific components of a person's immune system to fight diseases. This is usually done by stimulating or assisting one's immune system is attacking the offending agent - for instance, in the case of cancer - the target of immunotherapy will be cancer cells. Some types of immunotherapy are also called biologic therapy or biotherapy. One of the fundamental challenges that a living cell encounters are to accurately copy its genetic material to daughter cells during every single cell cycle. When this process goes haywire, genomic instability ensues, and genetic alterations ranging from nucleotide changes to chromosomal translocations and aneuploidy occur. Genomic instability arising out of DNA structural changes (indels, rearrangements, etc.,) can give rise to mutations predisposing to cancer. Cancer prevention refers to actions taken to mitigate the risk of getting cancer. The past decade has encountered an explosive rate of development of anticancer therapy ranging from standard chemotherapy to novel targeted small molecules that are nearly cancer specific, thereby reducing collateral damage. However, a new class of emerging therapy aims to train the body's defense system to fight against cancer. Termed as "cancer immunotherapy" is the new approach that has gained worldwide acceptance. It includes using antibodies that bind to and inhibit the function of proteins expressed by cancer cells or engineering and boosting the person's own T lymphocytes to target cancer. In this review, we summarized the recent advances and developments in cancer immunotherapy along with their shortcoming and challenges.
Collapse
Affiliation(s)
- Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ashikh Seethy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi; Department of Biochemistry, All India Institute of Medical Sciences, Guwahati, India
| | - Sunil Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Karthikeyan Pethusamy
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Tryambak Srivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Joyeeta Talukdar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Goura Kishor Rath
- Department of Radiation Oncology, DRBRAIRCH, All India Institute of Medical Sciences, New Delhi; Department of Radiation Oncology, NCI, All India Institute of Medical Sciences, Jhajjar, Haryana, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
28
|
Prognostic Factors of Second-line Immune Checkpoint Inhibitors in Patients With Advanced-stage Non-Small Cell Lung Cancer: A Multicenter, Retrospective Study. Am J Clin Oncol 2021; 44:356-360. [PMID: 34014843 DOI: 10.1097/coc.0000000000000828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICIs) targeting the programmed cell death receptor-1 and its ligand have achieved impressive success in treating patients with advanced-stage non-small cell lung cancer (NSCLC) after failed first-line cytotoxic chemotherapy. However, knowledge on clinical biomarkers that could help select patients who will respond well to second-line ICI therapy is limited. PATIENTS AND METHODS Medical records of patients with NSCLC treated with first-line platinum-based chemotherapy and subsequent second-line ICI were collected from 6 medical centers between January 2018 and June 2020. Clinical information, pathologic variables, and radiologic findings of the data collected were reviewed. The patients were followed up until the date of the last visit, the death of any cause, or the end of data recording (December 31, 2020). RESULTS A total of 181 patients with NSCLC were treated with second-line ICI following first-line platinum-based doublet chemotherapy. The median progression-free survival was 2.0 months (interquartile range, 1.0 to 5.5 mo), and the median overall survival was 12.0 months (interquartile range, 6.0 to 20.0 mo). Low body mass index (BMI) was independently associated with progression-free survival (odds ratio [OR], 0.826; 95% confidence interval [CI], 0.723-0.945; P=0.005). Similarly, a low BMI (OR, 0.839; 95% CI, 0.740-0.952; P=0.005) and a high number of metastatic organs (OR, 1.682; 95% CI, 1.156-2.448; P=0.007) were independently associated with the overall survival after second-line ICI therapy. CONCLUSION BMI and the number of metastatic sites were significantly associated with second-line ICI therapy outcomes in patients with NSCLC receiving first-line platinum-based chemotherapy.
Collapse
|
29
|
Zoi V, Galani V, Lianos GD, Voulgaris S, Kyritsis AP, Alexiou GA. The Role of Curcumin in Cancer Treatment. Biomedicines 2021; 9:biomedicines9091086. [PMID: 34572272 PMCID: PMC8464730 DOI: 10.3390/biomedicines9091086] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/19/2021] [Accepted: 08/23/2021] [Indexed: 12/24/2022] Open
Abstract
Curcumin is a polyphenol extracted from the rhizomes of the turmeric plant, Curcuma longa which has anti-inflammatory, and anticancer properties. Chronic inflammation is associated with the development of cancer. Curcumin acts on the regulation of various immune modulators, including cytokines, cyclooxygenase-2 (COX-2), and reactive oxygen species (ROS), which partly explains its anticancer effects. It also takes part in the downregulation of growth factors, protein kinases, oncogenic molecules and various signaling pathways, such as nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), c-Jun N-terminal kinase (JNK) and signal transducer and activator of transcription 3 (STAT3) signaling. Clinical trials of curcumin have been completed or are ongoing for various types of cancer. This review presents the molecular mechanisms of curcumin in different types of cancer and the evidence from the most recent clinical trials.
Collapse
Affiliation(s)
- Vasiliki Zoi
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Vasiliki Galani
- Department of Anatomy Histology-Embryology, School of Medicine, University of Ioannina, 45500 Ioannina, Greece;
| | - Georgios D. Lianos
- Department of Surgery, University Hospital of Ioannina, 45500 Ioannina, Greece;
| | - Spyridon Voulgaris
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
| | - Athanasios P. Kyritsis
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
| | - George A. Alexiou
- Neurosurgical Institute, School of Medicine, University of Ioannina, 45500 Ioannina, Greece; (V.Z.); (S.V.); (A.P.K.)
- Department of Neurosurgery, School of Medicine Ioannina, University of Ioannina, 45500 Ioannina, Greece
- Correspondence:
| |
Collapse
|
30
|
Sanaei MJ, Pourbagheri-Sigaroodi A, Kaveh V, Abolghasemi H, Ghaffari SH, Momeny M, Bashash D. Recent advances in immune checkpoint therapy in non-small cell lung cancer and opportunities for nanoparticle-based therapy. Eur J Pharmacol 2021; 909:174404. [PMID: 34363829 DOI: 10.1016/j.ejphar.2021.174404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/26/2021] [Accepted: 08/03/2021] [Indexed: 12/24/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer with the highest mortality rate and a poor 5-year survival rate. The majority of the cases are diagnosed in advanced stages when the disease has spread, which makes the tumor inoperable. Due to the antigenic essence of lung tumor cells, immunotherapy is a novel area and has exhibited remarkable results in this malignancy. Immune checkpoint inhibitors are inhibitory molecules that disrupt immune checkpoint signaling pathways whether in the immune cells or tumor cells. Tremelimumab and ipilimumab (CTLA-4 blockers), pembrolizumab and nivolumab (PD-1 blockers), and durvalumab, avelumab, and atezolizumab (PD-L1 blockers) are FDA-approved and improve the survival and objective response of NSCLC patients. Despite this, over-stimulation of the immune system via the immune checkpoint therapy is a double-edged sword that causes a spectrum of adverse events from moderate to life-threatening. Nanomedicine considerably impacts the way of diagnosis and treatment of tumors to overcome treatment-related challenges. Accordingly, nanoparticle-based immune checkpoint inhibitor therapy increases the local concentration of immune checkpoint inhibitors while reduces the side effects, which result in boosting the anti-tumor immunity against various types of malignancies, including NSCLC. The current review provides comprehensive information about immune checkpoint therapy in NSCLC, their efficacy, and their safety profile. Besides, recent advances in nanoparticle-based immune checkpoint therapy and its limitation are discussed.
Collapse
Affiliation(s)
- Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Kaveh
- Department of Medical Oncology and Hematology, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolghasemi
- Pediatric Congenital Hematologic Disorders Research Center, Mofid Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Romagnoli J, Tagliaferri L, Acampora A, Bianchi V, D'Ambrosio V, D'Aviero A, Esposito I, Hohaus S, Iezzi R, Lancellotta V, Maiolo E, Maiorano BA, Paoletti F, Peris K, Posa A, Preziosi F, Rossi E, Scaletta G, Schinzari G, Spagnoletti G, Tanzilli A, Scambia G, Tortora G, Valentini V, Maggiore U, Grandaliano G. Management of the kidney transplant patient with Cancer: Report from a Multidisciplinary Consensus Conference. Transplant Rev (Orlando) 2021; 35:100636. [PMID: 34237586 DOI: 10.1016/j.trre.2021.100636] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Cancer is the second most common cause of mortality and morbidity in Kidney Transplant Recipients (KTRs). Immunosuppression can influence the efficacy of cancer treatment and modification of the immunosuppressive regimen may restore anti-neoplastic immune responses improving oncologic prognosis. However, patients and transplant physicians are usually reluctant to modify immunosuppression, fearing rejection and potential graft loss. Due to the lack of extensive and recognised data supporting how to manage the immunosuppressive therapy in KTRs, in the context of immunotherapy, chemotherapy, radiotherapy and loco-regional treatments, a Consensus Conference was organised under the auspices of the European Society of Organ Transplantation and the Italian Society of Organ Transplantation. The conference involved a multidisciplinary group of transplant experts in the field across Europe. METHODS The overall process included a) the formulation of 12 specific questions based on the PICO methodology, b) systematic literature review and summary for experts for each question, c) a two-day conference celebration and the collection of experts' agreements. The conference was articulated in three sessions: "Immunosuppressive therapy and immunotherapy", "Systemic therapy", "Integrated Therapy", while the final experts' agreement was collected with a televoting procedure and defined according to the majority criterion. RESULTS Twenty-six European experts attended the conference and expressed their vote. A total of 14 statements were finally elaborated and voted. Strong agreement was found for ten statements, moderate agreement for two, moderate disagreement for one and uncertainty for the last one. CONCLUSIONS The consensus statements provide guidance to transplant physicians caring for kidney transplant recipients with cancer and indicate key aspects that need to be addressed by future clinical research.
Collapse
Affiliation(s)
- Jacopo Romagnoli
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Trapianti di Rene, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luca Tagliaferri
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.
| | - Anna Acampora
- Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Valentina Bianchi
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Trapianti di Rene, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia
| | - Viola D'Ambrosio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Andrea D'Aviero
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Ilaria Esposito
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. di Dermatologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Stefan Hohaus
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Roberto Iezzi
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. di Radiologia diagnostica e interventistica generale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Valentina Lancellotta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Elena Maiolo
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Brigida A Maiorano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy; Unità di Oncologia, Fondazione Casa Sollievo della Sofferenza IRCCS, San Giovanni Rotondo (FG), Italy
| | - Filippo Paoletti
- Facoltà di Medicina e Chirurgia, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ketty Peris
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. di Dermatologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Università Cattolica del Sacro Cuore, Dermatologia, Roma, Italy
| | - Alessandro Posa
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. di Radiologia diagnostica e interventistica generale, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Francesco Preziosi
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ernesto Rossi
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Oncologia Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giuseppe Scaletta
- Dipartimento della Salute della Donna, del Bambino e di Sanità Pubblica, UOC Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giovanni Schinzari
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Oncologia Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Gionata Spagnoletti
- Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Trapianti di Rene, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italia; Dipartimento di Chirurgie Specialistiche, Ch. Epato-Bilio-Pancreatica e Dei Trapianti di Fegato e Rene, Ospedale Pediatrico Bambino Gesù, IRCCS, Roma, Italy
| | - Alessandro Tanzilli
- Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giovanni Scambia
- Dipartimento Universitario di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento della Salute della Donna, del Bambino e di Sanità Pubblica, UOC Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giampaolo Tortora
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Oncologia Medica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Vincenzo Valentini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, U.O.C. Radioterapia Oncologica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Umberto Maggiore
- Dipartimento di Medicina e Chirurgia, Università di Parma, UO Nefrologia, Azienda-Ospedaliero di Parma, Parma, Italy
| | - Giuseppe Grandaliano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Roma, Italy; Dipartimento di Scienze Mediche e Chirurgiche, U.O.C. Nefrologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | | |
Collapse
|
32
|
Prasetya RA, Metselaar-Albers M, Engels F. Concomitant use of analgesics and immune checkpoint inhibitors in non-small cell lung cancer: A pharmacodynamics perspective. Eur J Pharmacol 2021; 906:174284. [PMID: 34174268 DOI: 10.1016/j.ejphar.2021.174284] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/20/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022]
Abstract
The invention of immunotherapy, such as immune checkpoint inhibitors (ICIs) for advanced-stage non-small cell lung cancer (NSCLC), has become a new standard of care for a defined group of NSCLC patients. However, the possible impacts of ICI interactions with analgesics for alleviating cancer-related pain are unclear and lack clinical evidence. Many studies have indicated that opioids detrimentally affect the immune system, possibly harming patients of ongoing immunotherapy. Opioids may repress the immune system in various ways, including impairing T cell function, upregulating immunosuppressor Treg cells, and interrupting intestinal microflora composition that disrupts the entire immune system. Furthermore, opioids can influence tumor progression and metastasis directly as opioid receptors are overexpressed in several types of NSCLC. In contrast, another analgesic acting on cyclooxygenase (COX) inhibition (i.e., NSAIDs) may be a candidate for adjuvant therapy since COX-2 is also expressed in the tumor cells of NSCLC patients. In addition, COX-2 is associated with tumor proliferation and metastasis. Therefore, both prospective and retrospective studies should confirm the advantages and disadvantages of the concurrent use of analgesics and ICIs in a clinical setting.
Collapse
Affiliation(s)
- Rahmad Aji Prasetya
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Department of Clinical Pharmacy, Akademi Farmasi Surabaya, Surabaya, Indonesia.
| | - Marjolein Metselaar-Albers
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Ferdi Engels
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
33
|
Darrigrand R, Pierson A, Rouillon M, Renko D, Boulpicante M, Bouyssié D, Mouton-Barbosa E, Marcoux J, Garcia C, Ghosh M, Alami M, Apcher S. Isoginkgetin derivative IP2 enhances the adaptive immune response against tumor antigens. Commun Biol 2021; 4:269. [PMID: 33649389 PMCID: PMC7921396 DOI: 10.1038/s42003-021-01801-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 02/05/2021] [Indexed: 11/25/2022] Open
Abstract
The success of cancer immunotherapy relies on the induction of an immunoprotective response targeting tumor antigens (TAs) presented on MHC-I molecules. We demonstrated that the splicing inhibitor isoginkgetin and its water-soluble and non-toxic derivative IP2 act at the production stage of the pioneer translation products (PTPs). We showed that IP2 increases PTP-derived antigen presentation in cancer cells in vitro and impairs tumor growth in vivo. IP2 action is long-lasting and dependent on the CD8+ T cell response against TAs. We observed that the antigen repertoire displayed on MHC-I molecules at the surface of MCA205 fibrosarcoma is modified upon treatment with IP2. In particular, IP2 enhances the presentation of an exon-derived epitope from the tumor suppressor nischarin. The combination of IP2 with a peptide vaccine targeting the nischarin-derived epitope showed a synergistic antitumor effect in vivo. These findings identify the spliceosome as a druggable target for the development of epitope-based immunotherapies.
Collapse
Affiliation(s)
- Romain Darrigrand
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - Alison Pierson
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - Marine Rouillon
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
- SATT Paris Saclay, Orsay, France
| | - Dolor Renko
- Université Paris-Saclay, CNRS, BioCIS, Châtenay-Malabry, France
| | - Mathilde Boulpicante
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France
| | - David Bouyssié
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Emmanuelle Mouton-Barbosa
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Julien Marcoux
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Camille Garcia
- Institut Jacques Monod, CNRS U7592 Université Paris Diderot, Paris, France
- Institut Pasteur, Unité de Spectrométrie de Masse pour la Biologie (MSBio), Centre de Ressources et Recherches Technologiques (C2RT), USR 2000 CNRS, Paris, France
| | - Michael Ghosh
- Department of Immunology, Interfaculty Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | - Mouad Alami
- Université Paris-Saclay, CNRS, BioCIS, Châtenay-Malabry, France
| | - Sébastien Apcher
- Université Paris-Saclay, Institut Gustave Roussy, Inserm, Immunologie des tumeurs et Immunothérapie, Villejuif, France.
| |
Collapse
|
34
|
Zhao L, Fu X, Han X, Yu Y, Ye Y, Gao J. Tumor mutation burden in connection with immune-related survival in uterine corpus endometrial carcinoma. Cancer Cell Int 2021; 21:80. [PMID: 33509222 PMCID: PMC7842022 DOI: 10.1186/s12935-021-01774-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/11/2021] [Indexed: 12/15/2022] Open
Abstract
Background UCEC is the most common gynecological malignancy in many countries, and its mechanism of occurrence and development is related to tumor mutation burden (TMB) and immune cell infiltration. Therefore, it is necessary to systematically explore the TMB-related gene profile in immune cells to improve the prognosis of UCEC. Methods We integrated TMB-related genes with basic clinical information of UCEC patients based on TCGA dataset. Differentially expressed genes (DEGs) were selected through differential expression screening, PPI, and enrichment analysis. Additionally, we analyzed the components of immune cell infiltration of the DEGs to obtain the differential immunity-related genes. A single factor and multifactor Cox regression analyses were conducted to establish new prognostic indicators of OS and DFS based on TMB-related immune genes. To further study the correlation between survival and immune cell infiltration, a Cox model based on these immune infiltration compositions was built. Using the clinical variables, we established nomograms for OS and DFS. Results 393 DEGs were significantly associated with clinical outcomes and the immune component in patients with UCEC. Gene Ontology (GO) and Kyoto Encyclopedia of Genes, Genomes (KEGG) pathway and protein-protein interaction network (PPI) analyses revealed the role of these genes and information on related pathways. Then, two prognostic models were established based on the differential immune genes for OS (GFAP and MX2) and DFS (MX2, GFAP, IGHM, FGF20, and TRAV21). In DFS, the differential immune genes were related to CD4+ T cell, CD8+ T cell, macrophage, and neutrophil (all P < 0.05). B cell and CD8+ T cell were independent prognostic factors from among the immune cell elements in UCEC. Finally, the risk scores of these models were combined with the clinical elements-based nomogram models, and the AUC values were all over 0.7. Conclusions Our results identified several clinically significant differential immune genes and established relevant prognostic models, providing a basis for the molecular analysis of TMB and immune cells in UCEC, and identified potential prognostic and immune-related genes for UCEC. We added clinical related conditions for further analysis to confirm the identity of the genes and clinical elements-based models.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Gynecology, Second Affiliated Hospital of Guizhou Medical University, Qiandongnan Second People's Hospital, Kaili, 556000, Guizhou, China
| | - Xueshu Fu
- Department of Gynecology, Huaian Maternity and Child Health Care Hospital, Huaian, 223002, Jiangsu, China
| | - Xiling Han
- Department of Obstetrics and Gynecology, Clinical Medical College of Yangzhou University, Yangzhou, 225001, Jiangsu, China
| | - Yanjun Yu
- Department of Gynecologic Oncology, Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Yaping Ye
- Department of Gynecologic Oncology, Dalian Medical University, Dalian, 116000, Liaoning, China
| | - Jun Gao
- Department of Obstetrics and Gynecology, Northern Jiangsu People's Hospital, 98 W Nantong Rd, Yangzhou, 225001, Jiangsu, China.
| |
Collapse
|
35
|
Guruprasad P, Lee YG, Kim KH, Ruella M. The current landscape of single-cell transcriptomics for cancer immunotherapy. J Exp Med 2021; 218:e20201574. [PMID: 33601414 PMCID: PMC7754680 DOI: 10.1084/jem.20201574] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
Immunotherapies such as immune checkpoint blockade and adoptive cell transfer have revolutionized cancer treatment, but further progress is hindered by our limited understanding of tumor resistance mechanisms. Emerging technologies now enable the study of tumors at the single-cell level, providing unprecedented high-resolution insights into the genetic makeup of the tumor microenvironment and immune system that bulk genomics cannot fully capture. Here, we highlight the recent key findings of the use of single-cell RNA sequencing to deconvolute heterogeneous tumors and immune populations during immunotherapy. Single-cell RNA sequencing has identified new crucial factors and cellular subpopulations that either promote tumor progression or leave tumors vulnerable to immunotherapy. We anticipate that the strategic use of single-cell analytics will promote the development of the next generation of successful, rationally designed immunotherapeutics.
Collapse
Affiliation(s)
- Puneeth Guruprasad
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Yong Gu Lee
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Ki Hyun Kim
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Marco Ruella
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
36
|
Vashi R, Patel BM. Roles of ARF tumour suppressor protein in lung cancer: time to hit the nail on the head! Mol Cell Biochem 2021; 476:1365-1375. [PMID: 33392921 DOI: 10.1007/s11010-020-03996-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022]
Abstract
Owing to its poor prognosis, the World Health Organization (WHO) lists lung cancer on top of the list when it comes to growing mortality rates and incidence. Usually, there are two types of lung cancer, small-cell lung cancer (SCLC) and non-small-cell lung cancer (NSCLC), which also includes adenocarcinoma, squamous cell carcinoma and large cell carcinomas. ARF, also known in humans as p14ARF and in the mouse as p19ARF, is a nucleolar protein and a member of INK4, a family of cyclin-independent kinase inhibitors (CKI). These genes are clustered on chromosome number 9p21 within the locus of CDKN2A. NSCLC has reported the role of p14ARF as a potential target. p14ARF has a basic mechanism to inhibit mouse double minute 2 protein that exhibits inhibitory action on p53, a phosphoprotein tumour suppressor, thus playing a role in various tumour-related activities such as growth inhibition, DNA damage, autophagy, apoptosis, cell cycle arrest and others. Extensive cancer research is ongoing and updated reports regarding the role of ARF in lung cancer are available. This article summarizes the available lung cancer ARF data, its molecular mechanisms and its associated signalling pathways. Attempts have been made to show how p14ARF functions in different types of lung cancer providing a thought to look upon ARF as a new target for treating the debilitating condition of lung cancer.
Collapse
Affiliation(s)
- Ruju Vashi
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India
| | - Bhoomika M Patel
- Institute of Pharmacy, Nirma University, Sarkhej-Gandhinagar Highway, Ahmedabad, Gujarat, 382 481, India.
| |
Collapse
|
37
|
Single-cell RNA sequencing reveals distinct tumor microenvironmental patterns in lung adenocarcinoma. Oncogene 2021; 40:6748-6758. [PMID: 34663877 PMCID: PMC8677623 DOI: 10.1038/s41388-021-02054-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/16/2021] [Accepted: 09/30/2021] [Indexed: 12/24/2022]
Abstract
Recent developments in immuno-oncology demonstrate that not only cancer cells, but also the tumor microenvironment can guide precision medicine. A comprehensive and in-depth characterization of the tumor microenvironment is challenging since its cell populations are diverse and can be important even if scarce. To identify clinically relevant microenvironmental and cancer features, we applied single-cell RNA sequencing to ten human lung adenocarcinomas and ten normal control tissues. Our analyses revealed heterogeneous carcinoma cell transcriptomes reflecting histological grade and oncogenic pathway activities, and two distinct microenvironmental patterns. The immune-activated CP²E microenvironment was composed of cancer-associated myofibroblasts, proinflammatory monocyte-derived macrophages, plasmacytoid dendritic cells and exhausted CD8+ T cells, and was prognostically unfavorable. In contrast, the inert N³MC microenvironment was characterized by normal-like myofibroblasts, non-inflammatory monocyte-derived macrophages, NK cells, myeloid dendritic cells and conventional T cells, and was associated with a favorable prognosis. Microenvironmental marker genes and signatures identified in single-cell profiles had progonostic value in bulk tumor profiles. In summary, single-cell RNA profiling of lung adenocarcinoma provides additional prognostic information based on the microenvironment, and may help to predict therapy response and to reveal possible target cell populations for future therapeutic approaches.
Collapse
|
38
|
Riccardo F, Barutello G, Petito A, Tarone L, Conti L, Arigoni M, Musiu C, Izzo S, Volante M, Longo DL, Merighi IF, Papotti M, Cavallo F, Quaglino E. Immunization against ROS1 by DNA Electroporation Impairs K-Ras-Driven Lung Adenocarcinomas . Vaccines (Basel) 2020; 8:vaccines8020166. [PMID: 32268572 PMCID: PMC7349290 DOI: 10.3390/vaccines8020166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/17/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is still the leading cause of cancer death worldwide. Despite the introduction of tyrosine kinase inhibitors and immunotherapeutic approaches, there is still an urgent need for novel strategies to improve patient survival. ROS1, a tyrosine kinase receptor endowed with oncoantigen features, is activated by chromosomal rearrangement or overexpression in NSCLC and in several tumor histotypes. In this work, we have exploited transgenic mice harboring the activated K-Ras oncogene (K-RasG12D) that spontaneously develop metastatic NSCLC as a preclinical model to test the efficacy of ROS1 immune targeting. Indeed, qPCR and immunohistochemical analyses revealed ROS1 overexpression in the autochthonous primary tumors and extrathoracic metastases developed by K-RasG12D mice and in a derived transplantable cell line. As proof of concept, we have evaluated the effects of the intramuscular electroporation (electrovaccination) of plasmids coding for mouse- and human-ROS1 on the progression of these NSCLC models. A significant increase in survival was observed in ROS1-electrovaccinated mice challenged with the transplantable cell line. It is worth noting that tumors were completely rejected, and immune memory was achieved, albeit only in a few mice. Most importantly, ROS1 electrovaccination was also found to be effective in slowing the development of autochthonous NSCLC in K-RasG12D mice.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Angela Petito
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Lidia Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Laura Conti
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Maddalena Arigoni
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Chiara Musiu
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Stefania Izzo
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Marco Volante
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging (IBB), Italian National Research Council (CNR), 10126 Torino, Italy;
| | - Irene Fiore Merighi
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
| | - Mauro Papotti
- Department of Oncology, University of Torino, 10043 Orbassano, Italy; (S.I.); (M.V.); (M.P.)
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
- Correspondence: (F.C.); (E.Q.); Tel.: +39-011670-6457 (F.C. & E.Q.)
| | - Elena Quaglino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (F.R.); (G.B.); (A.P.); (L.T.); (L.C.); (M.A.); (C.M.); (I.F.M.)
- Correspondence: (F.C.); (E.Q.); Tel.: +39-011670-6457 (F.C. & E.Q.)
| |
Collapse
|
39
|
Eriksson H, Smedby KE. Immune checkpoint inhibitors in cancer treatment and potential effect modification by age. Acta Oncol 2020; 59:247-248. [PMID: 32036733 DOI: 10.1080/0284186x.2020.1724329] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hanna Eriksson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Karin E. Smedby
- Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
- Division of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Korgaonkar N, Yadav KS. Understanding the biology and advent of physics of cancer with perspicacity in current treatment therapy. Life Sci 2019; 239:117060. [DOI: 10.1016/j.lfs.2019.117060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022]
|
41
|
NK Cells as Potential Targets for Immunotherapy in Endometriosis. J Clin Med 2019; 8:jcm8091468. [PMID: 31540116 PMCID: PMC6780982 DOI: 10.3390/jcm8091468] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/19/2022] Open
Abstract
Endometriosis is a common gynecological disease defined by the presence of endometrial-like tissue outside the uterus, most frequently on the pelvic viscera and ovaries, which is associated with pelvic pains and infertility. It is an inflammatory disorder with some features of autoimmunity. It is accepted that ectopic endometriotic tissue originates from endometrial cells exfoliated during menstruation and disseminating into the peritoneum by retrograde menstrual blood flow. It is assumed that the survival of endometriotic cells in the peritoneal cavity may be partially due to their abrogated elimination by natural killer (NK) cells. The decrease of NK cell cytotoxic activity in endometriosis is associated with an increased expression of some inhibitory NK cell receptors. It may be also related to the expression of human leukocyte antigen G (HLA-G), a ligand for inhibitory leukocyte immunoglobulin-like receptor subfamily B member 1 (LILRB1) receptors. The downregulated cytotoxic activity of NK cells may be due to inhibitory cytokines present in the peritoneal milieu of patients with endometriosis. The role of NK cell receptors and their ligands in endometriosis is also confirmed by genetic association studies. Thus, endometriosis may be a subject of immunotherapy by blocking NK cell negative control checkpoints including inhibitory NK cell receptors. Immunotherapies with genetically modified NK cells also cannot be excluded.
Collapse
|