1
|
Sharma A, Bhatia D. Programmable bionanomaterials for revolutionizing cancer immunotherapy. Biomater Sci 2024; 12:5415-5432. [PMID: 39291418 DOI: 10.1039/d4bm00815d] [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: 09/19/2024]
Abstract
Cancer immunotherapy involves a cutting-edge method that utilizes the immune system to detect and eliminate cancer cells. It has shown substantial effectiveness in treating different types of cancer. As a result, its growing importance is due to its distinct benefits and potential for sustained recovery. However, the general deployment of this treatment is hindered by ongoing issues in maintaining minimal toxicity, high specificity, and prolonged effectiveness. Nanotechnology offers promising solutions to these challenges due to its notable attributes, including expansive precise surface areas, accurate ability to deliver drugs and controlled surface chemistry. This review explores the current advancements in the application of nanomaterials in cancer immunotherapy, focusing on three primary areas: monoclonal antibodies, therapeutic cancer vaccines, and adoptive cell treatment. In adoptive cell therapy, nanomaterials enhance the expansion and targeting capabilities of immune cells, such as T cells, thereby improving their ability to locate and destroy cancer cells. For therapeutic cancer vaccines, nanoparticles serve as delivery vehicles that protect antigens from degradation and enhance their uptake by antigen-presenting cells, boosting the immune response against cancer. Monoclonal antibodies benefit from nanotechnology through improved delivery mechanisms and reduced off-target effects, which increase their specificity and effectiveness. By highlighting the intersection of nanotechnology and immunotherapy, we aim to underscore the transformative potential of nanomaterials in enhancing the effectiveness and safety of cancer immunotherapies. Nanoparticles' ability to deliver drugs and biomolecules precisely to tumor sites reduces systemic toxicity and enhances therapeutic outcomes.
Collapse
Affiliation(s)
- Ayushi Sharma
- Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura, Uttar Pradesh-281406, India.
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj 382355, Gandhinagar, India
| |
Collapse
|
2
|
Tellis JC, Wei B, Siu M, An L, Chan GK, Chen Y, Du X, Gazzard L, Hu B, Kiefer J, Kakiuchi-Kiyota S, Lainchbury M, Linehan JL, Luo X, Malhotra S, Mendonca R, Pang J, Ran Y, Sethuraman V, Seward E, Sneeringer C, Su D, Wang W, Wu P, Moffat JG, Heffron TP, Choo EF, Chan BK. Discovery of GNE-6893, a Potent, Selective, Orally Bioavailable Small Molecule Inhibitor of HPK1. ACS Med Chem Lett 2024; 15:1606-1614. [PMID: 39291002 PMCID: PMC11403726 DOI: 10.1021/acsmedchemlett.4c00319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) serves a key immunosuppressive role as a negative regulator of T-cell receptor (TCR) signaling. HPK1 loss-of-function is associated with augmentation of immune function and has demonstrated synergy with immune checkpoint inhibitors in syngeneic mouse cancer models. These data offer compelling evidence for the use of selective small molecule inhibitors of HPK1 in cancer immunotherapy. We identified a novel series of isoquinoline HPK1 inhibitors through fragment-based screening that displayed promising levels of biochemical potency and activity in functional cell-based assays. We used structure-based drug design to introduce key selectivity elements while simultaneously addressing pharmacokinetic liabilities. These efforts culminated in a molecule demonstrating subnanomolar biochemical inhibition of HPK1 and strong in vitro augmentation of TCR signaling in primary human T-cells. Further profiling of this molecule revealed excellent kinase selectivity (347/356 kinases <50% inhibition @ 0.1 μM), a favorable in vitro safety profile, and good projected human pharmacokinetics.
Collapse
Affiliation(s)
- John C Tellis
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - BinQing Wei
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Michael Siu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Le An
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Grace Kayan Chan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yong Chen
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Xiangnan Du
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lewis Gazzard
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Baihua Hu
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - James Kiefer
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Michael Lainchbury
- Charles River Laboratories, 8-9 Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Jonathan L Linehan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xifeng Luo
- Pharmaron Beijing Co., No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Sushant Malhotra
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Rohan Mendonca
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jodie Pang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yinqing Ran
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Vijay Sethuraman
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Eileen Seward
- Charles River Laboratories, 8-9 Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Chris Sneeringer
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Dian Su
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Weiru Wang
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ping Wu
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John G Moffat
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Timothy P Heffron
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Edna F Choo
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Bryan K Chan
- Genentech Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
3
|
Schirru M, Charef H, Ismaili KE, Fenneteau F, Zugaj D, Tremblay PO, Nekka F. Predicting efficacy assessment of combined treatment of radiotherapy and nivolumab for NSCLC patients through virtual clinical trials using QSP modeling. J Pharmacokinet Pharmacodyn 2024; 51:319-333. [PMID: 38493439 DOI: 10.1007/s10928-024-09903-0] [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: 11/07/2023] [Accepted: 02/05/2024] [Indexed: 03/19/2024]
Abstract
Non-Small Cell Lung Cancer (NSCLC) remains one of the main causes of cancer death worldwide. In the urge of finding an effective approach to treat cancer, enormous therapeutic targets and treatment combinations are explored in clinical studies, which are not only costly, suffer from a shortage of participants, but also unable to explore all prospective therapeutic solutions. Within the evolving therapeutic landscape, the combined use of radiotherapy (RT) and checkpoint inhibitors (ICIs) emerged as a promising avenue. Exploiting the power of quantitative system pharmacology (QSP), we undertook a study to anticipate the therapeutic outcomes of these interventions, aiming to address the limitations of clinical trials. After enhancing a pre-existing QSP platform and accurately replicating clinical data outcomes, we conducted an in-depth study, examining different treatment protocols with nivolumab and RT, both as monotherapy and in combination, by assessing their efficacy through clinical endpoints, namely time to progression (TTP) and duration of response (DOR). As result, the synergy of combined protocols showcased enhanced TTP and extended DOR, suggesting dual advantages of extended response and slowed disease progression with certain combined regimens. Through the lens of QSP modeling, our findings highlight the potential to fine-tune combination therapies for NSCLC, thereby providing pivotal insights for tailoring patient-centric therapeutic interventions.
Collapse
Affiliation(s)
- Miriam Schirru
- Laboratoire de recherche en pharmacométrie, Faculté de pharmacie, Université de Montréal, Montreal, Canada.
| | - Hamza Charef
- Laboratoire de recherche en pharmacométrie, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| | - Khalil-Elmehdi Ismaili
- Laboratoire de recherche en pharmacométrie, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| | - Frédérique Fenneteau
- Laboratoire de recherche en pharmacométrie, Faculté de pharmacie, Université de Montréal, Montreal, Canada
| | - Didier Zugaj
- Clinical Pharmacology, Syneos Health, Quebec, Quebec G1P 0A2, Canada
| | | | - Fahima Nekka
- Laboratoire de recherche en pharmacométrie, Faculté de pharmacie, Université de Montréal, Montreal, Canada
- Centre de recherches mathématiques (CRM), Université de Montréal, Montreal, Canada
- Centre for Applied Mathematics in Bioscience and Medicine (CAMBAM), McGill University, Montreal, Canada
| |
Collapse
|
4
|
Utkarsh K, Srivastava N, Kumar S, Khan A, Dagar G, Kumar M, Singh M, Haque S. CAR-T cell therapy: a game-changer in cancer treatment and beyond. Clin Transl Oncol 2024; 26:1300-1318. [PMID: 38244129 DOI: 10.1007/s12094-023-03368-2] [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: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024]
Abstract
In recent years, cancer has become one of the primary causes of mortality, approximately 10 million deaths worldwide each year. The most advanced, chimeric antigen receptor (CAR) T cell immunotherapy has turned out as a promising treatment for cancer. CAR-T cell therapy involves the genetic modification of T cells obtained from the patient's blood, and infusion back to the patients. CAR-T cell immunotherapy has led to a significant improvement in the remission rates of hematological cancers. CAR-T cell therapy presently limited to hematological cancers, there are ongoing efforts to develop additional CAR constructs such as bispecific CAR, tandem CAR, inhibitory CAR, combined antigens, CRISPR gene-editing, and nanoparticle delivery. With these advancements, CAR-T cell therapy holds promise concerning potential to improve upon traditional cancer treatments such as chemotherapy and radiation while reducing associated toxicities. This review covers recent advances and advantages of CAR-T cell immunotherapy.
Collapse
Affiliation(s)
- Kumar Utkarsh
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Namita Srivastava
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sachin Kumar
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Azhar Khan
- Faculty of Applied Science and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Gunjan Dagar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Mukesh Kumar
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mayank Singh
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Shabirul Haque
- Department of Autoimmune Diseases, Feinstein Institute for Medical Research, Northwell Health, 350, Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
5
|
Ma W, Hu J. Downregulated CDH3 is correlated with a better prognosis for LUAD and decreases proliferation and migration of lung cancer cells. Genes Genomics 2024; 46:713-731. [PMID: 38064156 DOI: 10.1007/s13258-023-01476-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 11/05/2023] [Indexed: 05/19/2024]
Abstract
BACKGROUND CDH3 is a glycoprotein with a single-span transmembrane domain that mediates cell-to-cell adhesion. Abnormal expression of CDH3 is associated with a poor prognosis in patients with breast, thyroid, colorectal carcinomas and glioblastoma. Soluble CDH3 in pleural effusions can be used as a marker for real-time monitoring of resistance to first- and second-generation EGFR-TKIs. The CDH3 mechanism underlying lung adenocarcinomas (LUADs) has not been established. OBJECTIVE This study analyzed the correlation between CDH3 expression and lung cancer prognosis and the effect of down-regulation CDH3 expression on the proliferation and migration of lung cancer cells. METHODS CDH3 expression was studied using the Oncomine, TIMER, PanglaoDB, and GEPIA databases. The effect of CDH3 on clinical prognosis was assessed with GEPIA, the PrognoScan database, and Kaplan-Meier plotter. The relationship between CDH3 to immune infiltrating cells was explored using TIMER and TISIDB. The function of CDH3 in lung cancer cell lines was determined by CCK-8 and wound healing assays in vitro. Furthermore, RNA sequencing was used to identify key signaling pathways and differentially-expressed genes. RESULTS LUAD tissues had higher CDH3 expression compared with normal tissues and were associated with worse overall survival in patients with LUAD. CDH3 expression had positive associations with infiltration of CD4 + T cells, Tregs and exhausted T cells, but negative associations with infiltration of B cells in patients with LUAD. CCK-8 and wound healing assays revealed that downregulation of CDH3 inhibited the proliferation and migration of cells. KEGG analysis revealed that the TGF-beta signaling pathways were demonstrated to be enriched pathways for genes negatively regulated by knockdown of CDH3. CONCLUSION CDH3 expression affects proliferation and migration of lung cancer cells and might serve as a potential prognostic marker in LUAD patients.
Collapse
Affiliation(s)
- Wanru Ma
- Department of Blood Transfusion, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, People's Republic of China
| | - Junhua Hu
- Department of Blood Transfusion, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, People's Republic of China.
| |
Collapse
|
6
|
Mang A, Zou W, Rolny V, Reck M, Cigoianu D, Schulze K, Holdenrieder S, Socinski MA, Shames DS, Wehnl B, Patil NS. Combined use of CYFRA 21-1 and CA 125 predicts survival of patients with metastatic NSCLC and stable disease in IMpower150. Tumour Biol 2024; 46:S177-S190. [PMID: 37545290 DOI: 10.3233/tub-230001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Patients with non-small cell lung cancer (NSCLC) and stable disease (SD) have an unmet clinical need to help guide early treatment adjustments. OBJECTIVE To evaluate the potential of tumor biomarkers to inform on survival outcomes in NSCLC SD patients. METHODS This post hoc analysis included 480 patients from the IMpower150 study with metastatic NSCLC, treated with chemotherapy, atezolizumab and bevacizumab combinations, who had SD at first CT scan (post-treatment initiation). Patients were stratified into high- and low-risk groups (overall survival [OS] and progression-free survival [PFS] outcomes) based on serum tumor biomarker levels. RESULTS The CYFRA 21-1 and CA 125 biomarker combination predicted OS and PFS in patients with SD. Risk of death was ~4-fold higher for the biomarker-stratified high-risk versus low-risk SD patients (hazard ratio [HR] 3.80; 95% confidence interval [CI] 3.02-4.78; p < 0.0001). OS in patients with the low- and high-risk SD was comparable to that in patients with the CT-defined partial response (PR; HR 1.10; 95% CI 0.898-1.34) and progressive disease (PD) (HR 1.05; 95% CI 0.621-1.77), respectively. The findings were similar with PFS, and consistent across treatment arms. CONCLUSIONS Biomarker testing shows potential for providing prognostic information to help direct treatment in NSCLC patients with SD. Prospective clinical studies are warranted.ClinicalTrials.gov: NCT02366143.
Collapse
Affiliation(s)
- Anika Mang
- Roche Diagnostics GmbH, Penzberg, Germany
| | - Wei Zou
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | | | - Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | | | - Katja Schulze
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | - Stefan Holdenrieder
- Institute of Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Munich, Germany
| | | | - David S Shames
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| | | | - Namrata S Patil
- Oncology Biomarkers Development, Genentech, San Francisco, CA, USA
| |
Collapse
|
7
|
Varshney K, Mazumder R, Rani A, Mishra R, Khurana N. Recent Research Trends against Skin Carcinoma - An Overview. Curr Pharm Des 2024; 30:2685-2700. [PMID: 39051578 DOI: 10.2174/0113816128307653240710044902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/29/2024] [Indexed: 07/27/2024]
Abstract
Skin cancer is a prevalent and sometimes lethal cancer that affects a wide range of people. UV radiation exposure is the main cause of skin cancer. Immunosuppression, environmental factors, and genetic predisposition are other contributing variables. Fair-skinned people and those with a history of sunburns or severe sun exposure are more likely to experience this condition. Melanoma, squamous cell carcinoma (SCC), and basal cell carcinoma (BCC) are the three main forms. Melanoma poses a bigger hazard because of its tendency for metastasis, while SCC and BCC have limited metastatic potential. Genetic mutations and changes to signalling pathways such as p53 and MAPK are involved in pathogenesis. Early diagnosis is essential, and molecular testing, biopsy, dermoscopy, and visual inspection can all help. In addition to natural medicines like curcumin and green tea polyphenols, treatment options include immunotherapy, targeted therapy, radiation, surgery, and chemotherapy. Reducing the incidence of skin cancer requires preventive actions, including sun protection and early detection programs. An overview of skin cancers, including their forms, pathophysiology, diagnosis, and treatment, highlighting herbal therapy, is given in this review.
Collapse
Affiliation(s)
- Kamya Varshney
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh 201306, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh 201306, India
| | - Anjna Rani
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh 201306, India
| | - Rashmi Mishra
- Department of Biotechnology, Noida Institute of Engineering and Technology, Greater Noida, Uttar Pradesh 201306, India
| | - Navneet Khurana
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
8
|
Xi J, Liu K, Peng Z, Dai X, Wang Y, Cai C, Yang D, Yan C, Li X. Toxic warhead-armed antibody for targeted treatment of glioblastoma. Crit Rev Oncol Hematol 2024; 193:104205. [PMID: 38036153 DOI: 10.1016/j.critrevonc.2023.104205] [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: 06/28/2023] [Revised: 10/31/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
Glioblastoma is a fatal intracranial tumor with a poor prognosis, exhibiting uninterrupted malignant progression, widespread invasion throughout the brain leading to the destruction of normal brain tissue and inevitable death. Monoclonal antibodies alone or conjugated with cytotoxic payloads to treat patients with different solid tumors showed effective. This treatment strategy is being explored for patients with glioblastoma (GBM) to obtain meaningful clinical responses and offer new drug options for the treatment of this devastating disease. In this review, we summarize clinical data (from pubmed.gov database and clinicaltrial.gov database) on the efficacy and toxicity of naked antibodies and antibody-drug conjugates (ADCs) against multiple targets on GBM, elucidate the mechanisms that ADCs act at the site of GBM lesions. Finally, we discuss the potential strategies for ADC therapies currently used to treat GBM patients.
Collapse
Affiliation(s)
- Jingjing Xi
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Kai Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhaolei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaolin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yulin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunyan Cai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Dejun Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chunmei Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiaofang Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
9
|
Singh S, Singh N, Baranwal M, Sharma S, Devi SSK, Kumar S. Understanding immune checkpoints and PD-1/PD-L1-mediated immune resistance towards tumour immunotherapy. 3 Biotech 2023; 13:411. [PMID: 37997595 PMCID: PMC10663421 DOI: 10.1007/s13205-023-03826-2] [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: 12/28/2022] [Accepted: 10/18/2023] [Indexed: 11/25/2023] Open
Abstract
Immunotherapy has emerged as a transformative approach in the treatment of various cancers, offering new hope for patients previously faced with limited treatment options. A cornerstone of cancer immunotherapy lies in targeting immune checkpoints, particularly the programmed cell death protein-1 (PD-1) and programmed death-ligand 1 (PD-L1) pathway. Immune checkpoints serve as crucial regulators of the immune response, preventing excessive immune activity and maintaining self-tolerance. PD-1, expressed on the surface of T cells, and its ligand PD-L1, expressed on various cell types, including cancer cells and immune cells, play a central role in this regulatory process. Although the success rate associated with these immunotherapies is very promising, most patients still show intrinsic or acquired resistance. Since the mechanisms related to PD-1/PD-L1 resistance are not well understood, an in-depth analysis is necessary to improve the success rate of anti-PD-1/PD-L1 therapy. Hence, here we provide an overview of PD-1, its ligand PD-L1, and the resistance mechanism towards PD-1/PD-L1. Furthermore, we have discussed the plausible solution to increase efficacy and clinical response. For the following research, joint endeavours of clinicians and basic scientists are essential to address the limitation of resistance towards immunotherapy.
Collapse
Affiliation(s)
- Sidhartha Singh
- School of Bioscience and Bioengineering, D Y Patil International University, Pune, Maharastra 411051 India
| | - Navneet Singh
- Department of Pulmonary Medicine, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012 India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, 147004 India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, 147004 India
| | - S. S. Kirthiga Devi
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037 India
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana 500037 India
| |
Collapse
|
10
|
Tojjari A, Saeed A, Sadeghipour A, Kurzrock R, Cavalcante L. Overcoming Immune Checkpoint Therapy Resistance with SHP2 Inhibition in Cancer and Immune Cells: A Review of the Literature and Novel Combinatorial Approaches. Cancers (Basel) 2023; 15:5384. [PMID: 38001644 PMCID: PMC10670368 DOI: 10.3390/cancers15225384] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/13/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
SHP2 (Src Homology 2 Domain-Containing Phosphatase 2) is a protein tyrosine phosphatase widely expressed in various cell types. SHP2 plays a crucial role in different cellular processes, such as cell proliferation, differentiation, and survival. Aberrant activation of SHP2 has been implicated in multiple human cancers and is considered a promising therapeutic target for treating these malignancies. The PTPN11 gene and functions encode SHP2 as a critical signal transduction regulator that interacts with key signaling molecules in both the RAS/ERK and PD-1/PD-L1 pathways; SHP2 is also implicated in T-cell signaling. SHP2 may be inhibited by molecules that cause allosteric (bind to sites other than the active site and attenuate activation) or orthosteric (bind to the active site and stop activation) inhibition or via potent SHP2 degraders. These inhibitors have anti-proliferative effects in cancer cells and suppress tumor growth in preclinical models. In addition, several SHP2 inhibitors are currently in clinical trials for cancer treatment. This review aims to provide an overview of the current research on SHP2 inhibitors, including their mechanism of action, structure-activity relationships, and clinical development, focusing on immune modulation effects and novel therapeutic strategies in the immune-oncology field.
Collapse
Affiliation(s)
- Alireza Tojjari
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Anwaar Saeed
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Arezoo Sadeghipour
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modarres University, Tehran P.O. Box 14115-175, Iran
| | - Razelle Kurzrock
- Department of Medicine, Genome Sciences and Precision Medicine Center, Medical College of Wisconsin Cancer Center, Milwaukee, WI 53226, USA
| | | |
Collapse
|
11
|
Rahman A, Janic B, Rahman T, Singh H, Ali H, Rattan R, Kazi M, Ali MM. Immunotherapy Enhancement by Targeting Extracellular Tumor pH in Triple-Negative Breast Cancer Mouse Model. Cancers (Basel) 2023; 15:4931. [PMID: 37894298 PMCID: PMC10605606 DOI: 10.3390/cancers15204931] [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: 08/25/2023] [Revised: 09/28/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Triple-negative breast cancer (TNBC), as one of the most aggressive forms of breast cancer, is characterized by a poor prognosis and a very low rate of disease-free and overall survival. In recent years, immunotherapeutic approaches targeting T cell checkpoint molecules, such as cytotoxic lymphocyte antigen-4 (CTLA-4), programmed death1 (PD-1) or its ligand, programmed death ligand 1 (PD-L1), have shown great potential and have been used to treat various cancers as single therapies or in combination with other modalities. However, despite this remarkable progress, patients with TNBC have shown a low response rate to this approach, commonly developing resistance to immune checkpoint blockade, leading to treatment failure. Extracellular acidosis within the tumor microenvironment (also known as the Warburg effect) is one of the factors preventing immune cells from mounting effective responses and contributing to immunotherapy treatment failure. Therefore, reducing tumor acidity is important for increasing cancer immunotherapy effectiveness and this has yet to be realized in the TNBC environment. In this study, the oral administration of sodium bicarbonate (NaHCO3) enhanced the antitumor effect of anti-PD-L1 antibody treatment, as demonstrated by generated antitumor immunity, tumor growth inhibition and enhanced survival in 4T1-Luc breast cancer model. Here, we show that NaHCO3 increased extracellular pH (pHe) in tumor tissues in vivo, an effect that was accompanied by an increase in T cell infiltration, T cell activation and IFN-γ, IL2 and IL12p40 mRNA expression in tumor tissues, as well as an increase in T cell activation in tumor-draining lymph nodes. Interestingly, these changes were further enhanced in response to combined NaHCO3 + anti-PD-L1 therapy. In addition, the acidic extracellular conditions caused a significant increase in PD-L1 expression in vitro. Taken together, these results indicate that alkalizing therapy holds potential as a new tumor microenvironment immunomodulator and we hypothesize that NaHCO3 can enhance the antitumor effects of anti-PD-L1 breast cancer therapy. The combination of these treatments may have an exceptional impact on future TNBC immunotherapeutic approaches by providing a powerful personalized medicine paradigm. Therefore, our findings have a great translational potential for improving outcomes in TNBC patients.
Collapse
Affiliation(s)
- Azizur Rahman
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Branislava Janic
- Department of Radiation Oncology, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Tasnim Rahman
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Harshit Singh
- Women’s Health Services, Henry Ford Hospital, Detroit, MI 48202, USA (R.R.)
| | - Haythem Ali
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | - Ramandeep Rattan
- Women’s Health Services, Henry Ford Hospital, Detroit, MI 48202, USA (R.R.)
| | - Mohsin Kazi
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Meser M. Ali
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| |
Collapse
|
12
|
Chen L, Wu Z, Guo J, Wang X, Zhao Z, Liang H, Zhang R, Deng J. Initial clinical and experimental analyses of ALDOA in gastric cancer, as a novel prognostic biomarker and potential therapeutic target. Clin Exp Med 2023; 23:2443-2456. [PMID: 36422738 DOI: 10.1007/s10238-022-00952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/15/2022] [Indexed: 11/25/2022]
Abstract
The effect of ALDOA, an important regulator of tumor metabolism and immune cell function, on gastric cancer (GC) immune infiltration has not been elucidated. Hence, we explored the feasibility of using ALDOA combined with immune molecular markers as novel prognostic or therapeutic targets for GC patients. Bioinformatic analyses were initially performed in multiple databases to assess the prognostic prediction values of ALDOA expression in GC. Subsequently, both ALDOA expression and the clinicopathological characteristics of a total of 114 GC patients who underwent curative gastrectomy were collected to demonstrate the potential association between ALDOA expression and the biological behaviors of GC. Next, the expression of ALDOA and its effect on prognosis were determined at the mRNA and protein levels, respectively, using tissue microarrays and cellular experiments. Subsequently, several molecular mechanisms were revealed based on elaborate analyses, indicating that ALDOA expression was potentially involved in the progression of GC and could be considered a promising biomarker for evaluating the prognosis of GC. High ALDOA expression was frequently found in GC cells and GC tissues at the mRNA and protein levels. Based on survival analysis, the expression of ALDOA indicated comparatively poor overall survival (OS) in GC and was identified as an independent prognostic predictor of GC. Correlation analysis showed that ALDOA expression had a positive association with lymph node metastasis in GC patients. Additionally, microRNA-1179 was found to play a key role in inhibiting the expression of ALDOA in the metabolic pathways of GC cells, which might disrupt the expression of various immune molecules and be detrimental to the prognosis of GC. ALDOA should be considered a promising molecular target for evaluating the prognosis of GC, owing to its potential role in immune regulation.
Collapse
Affiliation(s)
- Liqiao Chen
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Zizhen Wu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Jiamei Guo
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Xinyu Wang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Zhenzhen Zhao
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Rupeng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, People's Republic of China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, People's Republic of China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, People's Republic of China.
| |
Collapse
|
13
|
Aktar S, Islam F, Cheng T, Gamage SMK, Choudhury IN, Islam MS, Lu CT, Hamid FB, Ishida H, Abe I, Xie N, Gopalan V, Lam AK. Correlation between KRAS Mutation and CTLA-4 mRNA Expression in Circulating Tumour Cells: Clinical Implications in Colorectal Cancer. Genes (Basel) 2023; 14:1808. [PMID: 37761948 PMCID: PMC10530465 DOI: 10.3390/genes14091808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/07/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
Combination strategies of KRAS inhibition with immunotherapy in treating advanced or recurrent colorectal carcinoma (CRC) may need to be assessed in circulating tumour cells (CTCs) to achieve better clinical outcomes. This study aimed to investigate the genomic variations of KRAS in CTCs and matched CRC tissues and compared mRNA expression of KRAS and CTLA-4 between wild-type and KRAS-mutated CTCs and CRC tissues. Clinicopathological correlations were also compared. Six known mutations of KRAS were identified at both codon 12 and codon 13 (c.35G>T/G12V, c.35G>A7/G12D, c.35G>C/G12A, c.34G>A/G12S, c.38G>C/G13A, and c.38G>A/G13D). Three CTC samples harboured the identified mutations (16.7%; 3/18), while fifteen matched primary tumour tissues (65.2%, 15/23) showed the mutations. CTCs harbouring the KRAS variant were different from matched CRC tissue. All the mutations were heterozygous. Though insignificant, CTLA-4 mRNA expression was higher in patients carrying KRAS mutations. Patients harbouring KRAS mutations in CTCs were more likely to have poorly differentiated tumours (p = 0.039) and with lymph node metastasis (p = 0.027) and perineural invasion (p = 0.014). KRAS mutations in CTCs were also significantly correlated with overall pathological stages (p = 0.027). These findings imply the genetic basis of KRAS with immunotherapeutic target molecules based on a real-time platform. This study also suggests the highly heterogeneous nature of cancer cells, which may facilitate the assessment of clonal dynamics across a single patient's disease.
Collapse
Affiliation(s)
- Sharmin Aktar
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail 1902, Bangladesh
| | - Farhadul Islam
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tracie Cheng
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Sujani Madhurika Kodagoda Gamage
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Faculty of Health Sciences & Medicine, Bond University, Gold Coast, QLD 4229, Australia
| | - Indra Neil Choudhury
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Md Sajedul Islam
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Department of Biochemistry & Biotechnology, University of Barishal, Barishal 8254, Bangladesh
| | - Cu Tai Lu
- Department of Surgery, Gold Coast University Hospital, Gold Coast, QLD 4215, Australia;
| | - Faysal Bin Hamid
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Hirotaka Ishida
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Ichiro Abe
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Nan Xie
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
| | - Vinod Gopalan
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
| | - Alfred K. Lam
- Cancer Molecular Pathology, School of Medicine and Dentistry, Griffith University, Gold Coast, QLD 4222, Australia; (S.A.); (T.C.); (S.M.K.G.); (M.S.I.); (F.B.H.); (H.I.); (I.A.); (N.X.)
- Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD 4222, Australia;
- Pathology Queensland, Gold Coast University Hospital, Southport, QLD 4215, Australia
| |
Collapse
|
14
|
Fan Q, Chen H, Hu Y, Zhao B. Evaluation of uveitis events in real-world patients receiving immune checkpoint inhibitors based on the FAERS database. Cutan Ocul Toxicol 2023; 42:68-73. [PMID: 37130046 DOI: 10.1080/15569527.2023.2208661] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) have emerged as a novel class of drugs carrying a potential risk of uveitis. Due to the rarity, current knowledge on this safety issue is still incomplete. This study employed the post-marketing surveillance data to comprehensively describe and assess the uveitis events after the use of ICIs. METHODS Data between 2004 and 2021 were downloaded from the Food and Drug Administration Adverse Event Reporting System (FAERS), and the uveitis events reported for ICIs were identified and included in this study. Clinical details of these reports were collected and analyzed. Four data mining methods were utilized to investigate the potential associations between uveitis and different ICI regimens. RESULTS Overall, 461 uveitis cases after exposure to ICI therapies were reported. Melanoma (58.79%) was revealed as the most common indication for receiving ICIs. The median onset time of uveitis was 41 (interquartile range 18-91) days after ICI initiation. 9.54% of these cases resulted in disability. Data mining results showed 5 ICIs generated positive uveitis signals when used alone. Ipilimumab yielded the most noticeable uveitis signal with the highest reporting odds ratio (ROR = 6.73, 95% two-sided CI = 5.26, 8.60), proportional reporting ratio (PRR = 6.69, χ2=308.52), information component (IC = 2.74, IC025 = 2.14) and empirical Bayes geometric mean (EBGM = 6.66, EBGM05 = 5.42), followed by pembrolizumab, cemiplimab, nivolumab and atezolizumab. When nivolumab, pembrolizumab or atezolizumab was administrated together with ipilimumab, obviously stronger uveitis signal was detected than that for either of them. CONCLUSIONS This study provided an overview of the clinical features of ICI-related uveitis cases in the FAERS. Data mining results revealed that positive uveitis signals commonly existed within this drug class, but signal strength varied among ICIs. When ICIs were used in a combined way, uveitis signals became obviously stronger. Therefore, early ophthalmic monitoring is important when applying ICIs to patients, especially those with a tendency for uveitis, such as melanoma patients.
Collapse
Affiliation(s)
- Qianqian Fan
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Huan Chen
- Department of Ophthalmology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yang Hu
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Bin Zhao
- Department of Pharmacy, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Chen D, Zhang H, Zhao L, Liu X, Xue S, Wu P, Jiang H. Prognostic value of RILPL2 and its correlation with tumor immune microenvironment and glycolysis in non-small cell lung cancer. Cell Cycle 2023; 22:841-857. [PMID: 36536539 PMCID: PMC10026870 DOI: 10.1080/15384101.2022.2159203] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Rab-interacting lysosomal protein - like 2 (RILPL2) has been reported to be associated with prognosis and tumor biological functions in breast cancer and endometrial carcinoma. However, its expression and functional role in non-small cell lung cancer (NSCLC) remain unclear. The expression and clinical data of lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC) were downloaded from the TCGA database. The expression of RILPL2 in NSCLC cell lines was verified by the Western blot. We used online databases and bioinformatics analysis tools to explore its prognostic value, potential biological functions, and correlations with tumor immune microenvironment.The expression of RILPL2 was significantly lower in NSCLC compared with adjacent normal tissues. Low RILPL2 expression was associated with worse overall survival (OS) in NSCLC. The GO analysis showed RILPL2 was comprehensively involved in immune activity. RILPL2 expression was significantly positively correlated with the infiltration levels of B cells, CD8+T cells, CD4+T cells, macrophages, neutrophils, dendritic cells (P < 0.001), and it was also significantly positively correlated with programmed cell death ligand 1 (PD-L1/CD274) (P < 0.001). High RILPL2 expression could predict better immunotherapy response and prognosis in the immunotherapy cohort. The GSEA analysis showed low RILPL2 expression was associated with glycolysis process in LUAD, which was verified in vitro.These results showed RILPL2 expression was correlated with prognosis, tumor microenvironment, and immunotherapy response in NSCLC. Besides, RILPL2 may regulate glycolysis in LUAD.
Collapse
Affiliation(s)
- Dongfang Chen
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Hongyan Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Lifang Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Xueqing Liu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Shan Xue
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Peiling Wu
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| | - Handong Jiang
- Department of Respiratory and Critical Care Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
16
|
Zhang JJ, Zhang Y, Chen Q, Chen QN, Yang X, Zhu XL, Hao CY, Duan HB. A Novel Prognostic Marker and Therapeutic Target Associated with Glioma Progression in a Tumor Immune Microenvironment. J Inflamm Res 2023; 16:895-916. [PMID: 36883185 PMCID: PMC9985882 DOI: 10.2147/jir.s398775] [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: 11/28/2022] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
Background Immune microenvironment serves a vital role in glioma progression, and a large number of studies have found that tumor progression can be reduced to some extent by modulating the immune process in tumors. Materials and Methods ImmuneScore of each sample in CGGA datasets were calculated with Estimate R package, and samples were grouped by median ImmuneScore values for differential analysis to obtain immune microenvironment differential genes. We further conducted survival analysis, ROC curve analysis, independent prognostic analysis, and clinical correlation analysis on glioma sample genes in CGGA to obtain glioma prognostic genes, and then identified their intersection with immune microenvironment DEGs by Venn tool. The GEPIA and UALCAN databases were used to verify the differential expression of intersecting genes in the glioma and normal brain and to identify our target gene. After validation of their prognostic value, we constructed a nomogram to calculate the risk score and to estimate the accuracy of prognostic model. We mined co-expression genes, enriched functions and pathways, and correlations to immune cell infiltration of unigene with an online database. Finally, we verified the differential expression of FCGBP in glioma by immunohistochemical staining. Results We finally selected Fc fragment of IgG-binding protein (FCGBP) as our study gene. The prognostic values of FCGBP were validated by a series of analyses. Immunohistochemical staining showed that FCGBP expression increased in gliomas and was up-regulated with the progression of glioma grade. Conclusion As a key unigene in glioma progression, FCGBP contributes to the regulation of immune microenvironment and has the potential to be a prognostic biomarker and immune targets.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Yu Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qian Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Qi-Ning Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xin Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Xiao-Lin Zhu
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chun-Yan Hao
- Department of Geriatrics, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Hu-Bin Duan
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| |
Collapse
|
17
|
Banerjee S, Nahar U, Dahiya D, Mukherjee S, Dey P, Gupta R, Radotra B, Sachdeva N, Sood A, Bhadada SK, Bhansali A. Role of cytotoxic T cells and PD-1 immune checkpoint pathway in papillary thyroid carcinoma. Front Endocrinol (Lausanne) 2022; 13:931647. [PMID: 36518249 PMCID: PMC9742369 DOI: 10.3389/fendo.2022.931647] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022] Open
Abstract
Background Lymphocytic thyroiditis (LT) is frequently seen in the tumor microenvironment (TME) of papillary thyroid carcinomas (PTCs). However, the characteristic of these tumor-infiltrating lymphocytes (TILs) is not well understood. Objective We aim to define the TME of PTC cases by characterizing the TILs. Design This is a cross-sectional observational study. Patients We enrolled 29 PTC (23 having concurrent LT), 14 LT, and 13 hyperplastic nodules with LT (HN) patients from January 2016 to December 2020. Measurements Immunohistochemical (IHC) expression of CD8, FoxP3, PD-1, and PD-L1 was studied in PTC with LT and compared with HN. PD-1 and PD-L1 expression was correlated at the mRNA level by quantitative real-time PCR. Immunophenotyping of TILs was done in FNAC samples of PTC and LT by flow cytometry. Results IHC revealed the presence of CD8+ cytotoxic T lymphocytes (CTLs) and FoxP3+ T regulatory cells (Tregs) in 83% and 52% of PTC with LT cases, respectively. Flow cytometric analysis of the PTC samples revealed a significant abundance of CTL compared with Treg and a higher CTL with lower Treg counts compared with LT. On IHC, PD-1 positivity was noted in 56.5% of PTC with LT cases, while intermediate PD-L1 positivity was found in 70% of the cases. There was a significant upregulation of PD-1 mRNA in PTC with LT. A significant correlation was noted with PD-L1 expression with lymph node metastasis and presence of Treg cells. Conclusions Increased expression of PD-1 and PD-L1 in the TME of PTC may provide a potential molecular mechanism for tumor survival despite the predominance of CTLs, possibly through their inactivation or exhaustion.
Collapse
Affiliation(s)
- Sohini Banerjee
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Uma Nahar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Soham Mukherjee
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pranab Dey
- Department of Cytology and Gynaecological Pathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Rijuneeta Gupta
- Department of Otolaryngology (ENT), Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bishan Radotra
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Naresh Sachdeva
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashwani Sood
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
18
|
Shen T, Cai W, Li T, Yu D, Ren C, Yu J. Impact of primary site on survival in patients with nasopharyngeal carcinoma from 2004 to 2015. Front Surg 2022; 9:1001849. [PMID: 36406355 PMCID: PMC9671952 DOI: 10.3389/fsurg.2022.1001849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022] Open
Abstract
Background Nasopharynx carcinoma (NPC) is the most common malignant tumor of the nasopharynx. Many studies have shown some factors related with the prognosis of NPC patients. Our study aims to evaluate the differences of prognosis between initial and second primary NPC. Material and methods The Surveillance, Epidemiology, and End Results (SEER) program was used to perform the population-based analysis in NPC patients who were newly diagnosed between 2004 and 2015. Kaplan-Meier and Cox regressions were used to evaluate the effects of primary site on the overall survival (OS), as well as the cancer-specific survival (CSS). Results Our study included 5,012 NPC patients: 4,474 initial primary NPC patients and 5,38 s primary NPC patients. Significant differences were observed in sex, age at diagnosis, race, median household income, histological type, American Joint Committee on Cancer (AJCC) stage, N-stage, radiation treatment and chemotherapy between patients with initial and second NPC (P < 0.05). Moreover, the patients with second NPC had longer survival months. In addition, radiation and chemotherapy were recommended both in first and second primary NPC patients. Conclusion Worse prognosis was observed in patients with second primary NPC compared with those with primary NPC in all subgroups of AJCC stage and age at diagnosis.
Collapse
Affiliation(s)
- Tianyi Shen
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Wenting Cai
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Tingting Li
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Donghui Yu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Chengda Ren
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jing Yu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University, School of Medicine, Shanghai, China
- Department of Ophthalmology, The Third People’s Hospital of Bengbu, Bengbu, China
- Correspondence: Jing Yu
| |
Collapse
|
19
|
Li X, Lu D, Zhang Z, Zhang Y, Wang J, Hu Y. Prognostic value of plasma D-dimer levels in advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors: a retrospective study. J Thorac Dis 2022; 14:4125-4135. [PMID: 36389301 PMCID: PMC9641356 DOI: 10.21037/jtd-22-1363] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/19/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Plasma D-dimer is of great significance for the clinical exclusion of tumor-related thrombosis. Previous studies have shown its predictive role in non-small cell lung cancer (NSCLC) treated with chemotherapy. However, whether pretreatment D-dimer could predict the efficacy and prognosis in NSCLC patients treated with immune checkpoint inhibitors (ICIs) remains unclear. METHODS Advanced NSCLC patients treated with ICIs at the Chinese PLA General Hospital between January 2015 and March 2019 were enrolled. Patients were divided into a pretreatment normal D-dimer group (≤0.5 µg/mL) and high D-dimer group (>0.5 µg/mL). Optimization-based approach was applied to balance baseline covariates between the 2 groups, including age, sex, histological type, smoking history, stage, Eastern Cooperative Oncology Group Performance Status (ECOG PS), lines of treatment, ICI drugs, brain metastasis, treatment type, and D-dimer levels. Kaplan-Meier analysis and Cox proportional hazards model were used for analyzing survival data, including progression-free survival (PFS, the time from initial ICI treatment to PD or death), overall survival (OS, the time between initial ICI treatment and death), and hazard ratio (HR). Follow-up of all patients was performed by searching electronic medical records and counseling telephone. The follow-up cut-off date was July 6, 2020. RESULTS This study included 277 advanced NSCLC patients. Among the enrolled patients, 23.1% were female, 64.6% had non-squamous cell lung cancer, and 79.4% were stage IV. Univariate and multivariate analysis showed that pretreatment high D-dimer levels were independently associated with shortened PFS and OS (P<0.01). Subgroup analysis confirmed that pretreatment high D-dimer levels were associated with poor prognosis in most subsets. After balancing baseline covariates between the high D-dimer group and normal D-dimer group, the results indicated that patients with pretreatment high D-dimer levels had significantly shorter PFS [median: 6.4 vs. 11.5 months; HR, 1.70; 95% confidence ratio (CI): 1.25-2.37; P<0.001] and OS (median: 12.7 vs. 30.4 months; HR, 2.29; 95% CI: 1.54-3.41; P<0.001) than those with pretreatment normal D-dimer levels. CONCLUSIONS Pretreatment plasma D-dimer could serve as a convenient prognostic biomarker for advanced NSCLC patients receiving ICI treatment. Patients with pretreatment high D-dimer levels may have poor PFS and OS.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Di Lu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Zhibo Zhang
- Department of Cardiothoracic Surgery, The 78th Group Army Hospital of Chinese PLA, Mudanjiang, China
| | - Yuning Zhang
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China;,Medical School of Chinese PLA, Beijing, China
| | - Jinliang Wang
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yi Hu
- Department of Medical Oncology, Senior Department of Oncology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
TIMP2 is associated with prognosis and immune infiltrates of gastric and colon cancer. Int Immunopharmacol 2022; 110:109008. [DOI: 10.1016/j.intimp.2022.109008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023]
|
21
|
Lu Y, Yu J, Dong Q, Du Y, Liang Z. DOCK4 as a Potential Biomarker Associated with Immune Infiltration in Stomach Adenocarcinoma: A Database Analysis. Int J Gen Med 2022; 15:6127-6143. [PMID: 35846794 PMCID: PMC9286484 DOI: 10.2147/ijgm.s357096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/29/2022] [Indexed: 12/24/2022] Open
Abstract
Purpose The involvement of dedicator for cytokinesis 4 (DOCK4), a guanine nucleotide exchange factor for Rac1, in immune infiltration in stomach adenocarcinoma (STAD) remains unclear. Methods The UALCAN database was used to analyze the expression of the DOCK family. The Kaplan–Meier method and Gene Expression Profiling Interactive Analysis (GEPIA) databases were used to assess the prognostic value of the DOCK family in STAD. Furthermore, the correlation between expression of DOCK4 as well as other immune-related marker genes and tumor immune infiltration in STAD was explored using the TIMER and GEPIA websites. Subsequently, the relationship between DOCK4 expression and clinical characteristics was verified using the UALCAN database. Finally, DOCK4 mutation was analyzed via the TIMER2.0 and cBioPortal databases and the DOCK4 protein-protein interaction networks were constructed using the GeneMANIA and STRING websites. Results DOCK4 was found to be a new prognostic biomarker in STAD. DOCK4 expression in tumors was thoroughly evaluated relative to paracancerous tissues; overexpression of DOCK4 had a negative impact on the prognosis of patients with STAD. DOCK4 was found to be significantly associated with tumor immune infiltration in STAD. Conclusion In summary, DOCK4 is a potential regulator of the recruitment and regulation of immune-infiltrating cells, thus serving as a valuable prognostic biomarker in STAD.
Collapse
Affiliation(s)
- Yi Lu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Jiaxi Yu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Qiuping Dong
- Department of Cancer Cell Biology, Tianjin's Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, People's Republic of China
| | - Yan Du
- Department of Operating Theatre, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Zheng Liang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| |
Collapse
|
22
|
High Expression of TACC3 Is Associated with the Poor Prognosis and Immune Infiltration in Lung Adenocarcinoma Patients. DISEASE MARKERS 2022; 2022:8789515. [PMID: 35855850 PMCID: PMC9288335 DOI: 10.1155/2022/8789515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 06/17/2022] [Indexed: 12/02/2022]
Abstract
Background Lung adenocarcinoma (LUAD) has been recognized as one of the commonest aggressive malignant tumors occurring in humans. The transforming acidic coiled-coil-containing protein 3 (TACC3) seems to be a probable prognostic marker and treatment target for non-small-cell lung cancer (NSCLC). Nevertheless, there exist no reports on the association between TACC3 and immunotherapy or other therapeutic interventions in LUAD. Methods Premised on the data accessed from The Cancer Genome Atlas- (TCGA-) LUAD, we carried out bioinformatics analysis. The TACC3 expression in LUAD was analyzed utilizing the GEPIA. A survival module was constructed to evaluate the effect of TACC3 on the survival of patients with LUAD. Logistic regression was undertaken to examine the relationship between TACC3 expression and clinical factors. Protein-protein interaction analysis was performed in the GeneMANIA database, and enrichment analysis and identification of predicted signaling pathways were performed using Gene Ontology and Kyoto Encyclopedia of Genes. Additionally, the Cox regression was used to assess the clinicopathologic features linked to the overall survival in TCGA patients. Lastly, we investigated the link between TACC3 and tumor-infiltrating immune cells (TIICs) through CIBERSORT and the “Correlation” module of GEPIA. The association between TACC3 gene expression and drug response was analyzed using the CellMiner database to predict drug sensitivity. Results The outcomes illustrated that TACC3 was upregulated and considerably correlated with dismal prognosis in LUAD patients. Moreover, the multivariate Cox regression analysis depicted TACC3 as an independent prognostic marker in LUAD patients. It was also revealed that the expression of TACC3 was related to clinical stage (P = 0.014), age (P = 0.002), and T classification (P ≤ 0.018). Moreover, we discovered that the expression of TACC3 was considerably linked to a wide range of TIICs, especially the T cells and NK cells. Single-cell results found that TACC3 was mainly expressed in the immune cells (especially tprolif cells) and malignant cells. TACC3 gene expression was positively correlated with TMB and MSI, and TACC3 may provide a prediction of the efficacy of immunotherapy. Moreover, the correlation analysis between TACC3 gene expression and immune checkpoint gene expression revealed that TACC3 may coordinate the activities of these ICP genes in different signal transduction pathways. TACC3 is related to biological progress (BP), cellular component (CC), and molecular function (MF). The pathways involved in the interaction network involving TACC3 include nonhomologous end-joining, RNA transport, pantothenate and CoA biosynthesis, homologous recombination, and nucleotide excision repair. Furthermore, we investigated the association between the expression of TACC3 and the use of antitumor drugs, and TACC3 was positively correlated with response to most drugs. Conclusion The findings from this research offer robust proof that the expression of TACC3 could be a prognostic marker correlated with TIICs in LUAD. TACC3 can also provide new ideas for immunotherapy as a potential therapeutic target.
Collapse
|
23
|
Wu G, Chen M, Ren H, Sha X, He M, Ren K, Qi J, Lin F. AP3S1 is a Novel Prognostic Biomarker and Correlated With an Immunosuppressive Tumor Microenvironment in Pan-Cancer. Front Cell Dev Biol 2022; 10:930933. [PMID: 35874816 PMCID: PMC9304770 DOI: 10.3389/fcell.2022.930933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Adaptor-related protein complex 3, sigma one subunit (AP3S1) is one of the encoding subunits of the adaptor complex AP-3. However, its role in various tumor types and relationship with the tumor immune microenvironment (TIME) remains unclear.Methods: AP3S1 expression was analyzed using datasets from The Cancer Genome Atlas, Genotype-Tissue Expression, UALCAN, and HPA databases. Then, we performed a systematic analysis of the genetic alterations, clinical features, and prognostic value of AP3S1 in pan-cancer. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were used to identify the signaling pathways associated with AP3S1. The correlation between immune cell infiltration and AP3S1 expression was analyzed using immune cell infiltration data from the ImmuCellAI, TIMER2, and a previous study. Finally, we analyzed the association of AP3S1 with tumor mutational burden (TMB), microsatellite instability (MSI), and immune-related genes.Results: We found AP3S1 overexpression in most tumors and a significant association with low survival rates. GSEA and GSVA results show that AP3S1 is involved in tumor progression and associated with immune pathways in different tumor types. We also found that AP3S1 expression was positively correlated with the level of infiltration of immunosuppressive cells (tumor-associated macrophages, cancer-associated fibroblasts, Tregs) and negatively correlated with immune killer cells, including NK cells and CD8+ T cells, in pan-cancer. The expression of AP3S1 could affect TMB and MSI in various cancers. In addition, AP3S1 was positively correlated with most immunosuppressive genes, including PD-1, PD-L1, CTLA4, LAG3 and TIGIT in most cancer types.Conclusion: Our study reveals that AP3S1 is a potential pan-cancer oncogene and plays an essential role in tumorigenesis and cancer immunity. Elevated expression of AP3S1 indicates an immunosuppressive microenvironment and can be used as a potential prognostic biomarker and a target for immunotherapy.
Collapse
Affiliation(s)
- Gujie Wu
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Mianxiong Chen
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Hefei Ren
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Xinyu Sha
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Kuan Ren
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Juntao Qi
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
- *Correspondence: Juntao Qi, ; Feng Lin,
| | - Feng Lin
- Department of Urology, Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
- *Correspondence: Juntao Qi, ; Feng Lin,
| |
Collapse
|
24
|
Wang XF, Lei W, Liu CM, Yang J, Zhu YH. BOLA3 is a prognostic-related biomarker and correlated with immune infiltrates in lung adenocarcinoma. Int Immunopharmacol 2022; 107:108652. [DOI: 10.1016/j.intimp.2022.108652] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 02/07/2022] [Accepted: 02/20/2022] [Indexed: 12/20/2022]
|
25
|
He ZX, Zhao SB, Fang X, E JF, Fu HY, Song YH, Wu JY, Pan P, Gu L, Xia T, Liu YL, Li ZS, Wang SL, Bai Y. Prognostic and Predictive Value of BGN in Colon Cancer Outcomes and Response to Immunotherapy. Front Oncol 2022; 11:761030. [PMID: 35096572 PMCID: PMC8790701 DOI: 10.3389/fonc.2021.761030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background Colon cancer is one of the most frequent malignancies and causes high mortality worldwide. Exploring the tumor-immune interactions in the tumor microenvironment and identifying new prognostic and therapeutic biomarkers will assist in decoding the novel mechanism of tumor immunotherapy. BGN is a typical extracellular matrix protein that was previously validated as a signaling molecule regulating multiple processes of tumorigenesis. However, its role in tumor immunity requires further investigation. Methods The differentially expressed genes in three GEO datasets were analyzed, and BGN was identified as the target gene by intersection analysis of PPIs. The relevance between clinical outcomes and BGN expression levels was evaluated using data from the GEO database, TCGA and tissue microarray of colon cancer samples. Univariable and multivariable Cox regression models were conducted for identifying the risk factors correlated with clinical prognosis of colon cancer patients. Next, the association between BGN expression levels and the infiltration of immune cells as well as the process of the immune response was analyzed. Finally, we predicted the immunotherapeutic response rates in the subgroups of low and high BGN expression by TIS score, ImmuCellAI and TIDE algorithms. Results BGN expression demonstrated a statistically significant upregulation in colon cancer tissues than in normal tissues. Elevated BGN was associated with shorter overall survival as well as unfavorable clinicopathological features, including tumor size, serosa invasion and length of hospitalization. Mechanistically, pathway enrichment and functional analysis demonstrated that BGN was positively correlated with immune and stromal scores in the TME and primarily involved in the regulation of immune response. Further investigation revealed that BGN was strongly expressed in the immunosuppressive phenotype and tightly associated with the infiltration of multiple immune cells in colon cancer, especially M2 macrophages and induced Tregs. Finally, we demonstrated that high BGN expression presented a better immunotherapeutic response in colon cancer patients. Conclusion BGN is an encouraging predictor of diagnosis, prognosis and immunotherapeutic response in patients with colon cancer. Assessment of BGN expression represents a novel approach with great promise for identifying patients who may potentially benefit from immunotherapy.
Collapse
Affiliation(s)
- Zi-Xuan He
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Xue Fang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Ji-Fu E
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Hong-Yu Fu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yi-Hang Song
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Jia-Yi Wu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Peng Pan
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Lun Gu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Tian Xia
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yi-Long Liu
- College of Basic Medicine Sciences, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| |
Collapse
|
26
|
Chan BK, Seward E, Lainchbury M, Brewer TF, An L, Blench T, Cartwright MW, Chan GKY, Choo EF, Drummond J, Elliott RL, Gancia E, Gazzard L, Hu B, Jones GE, Luo X, Madin A, Malhotra S, Moffat JG, Pang J, Salphati L, Sneeringer CJ, Stivala CE, Wei B, Wang W, Wu P, Heffron TP. Discovery of Spiro-azaindoline Inhibitors of Hematopoietic Progenitor Kinase 1 (HPK1). ACS Med Chem Lett 2021; 13:84-91. [PMID: 35059127 PMCID: PMC8762754 DOI: 10.1021/acsmedchemlett.1c00473] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/01/2021] [Indexed: 01/16/2023] Open
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is implicated as a negative regulator of T-cell receptor-induced T-cell activation. Studies using HPK1 kinase-dead knock-in animals have demonstrated the loss of HPK1 kinase activity resulted in an increase in T-cell function and tumor growth inhibition in glioma models. Herein, we describe the discovery of a series of small molecule inhibitors of HPK1. Using a structure-based drug design approach, the kinase selectivity of the molecules was significantly improved by inducing and stabilizing an unusual P-loop folded binding mode. The metabolic liabilities of the initial 7-azaindole high-throughput screening hit were mitigated by addressing a key metabolic soft spot along with physicochemical property-based optimization. The resulting spiro-azaindoline HPK1 inhibitors demonstrated improved in vitro ADME properties and the ability to induce cytokine production in primary human T-cells.
Collapse
Affiliation(s)
- Bryan K. Chan
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States,
| | - Eileen Seward
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Michael Lainchbury
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Thomas F. Brewer
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Le An
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Toby Blench
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Matthew W. Cartwright
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Grace Ka Yan Chan
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Edna F. Choo
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jason Drummond
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Richard L. Elliott
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Emanuela Gancia
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Lewis Gazzard
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Baihua Hu
- Pharmaron
Beijing Co, No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Graham E. Jones
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Xifeng Luo
- Pharmaron
Beijing Co, No. 6 Tai He Road, BDA, Beijing 100176, P.R. China
| | - Andrew Madin
- Charles
River Laboratories, 8-9
Spire Green, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Sushant Malhotra
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John G. Moffat
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jodie Pang
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Laurent Salphati
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | | | - Craig E. Stivala
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Binqing Wei
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Weiru Wang
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ping Wu
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Timothy P. Heffron
- Genentech
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
27
|
Geng L, Chen S, Gong Y, Zhou Y, Yang H, Tang L. Tumor Endothelial Marker TEM7 is a Prognostic Biomarker and Correlating with Immune Infiltrates in Gastric Cancer. Int J Gen Med 2021; 14:10155-10171. [PMID: 34992436 PMCID: PMC8711244 DOI: 10.2147/ijgm.s347010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 12/09/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction Tumor endothelial marker 7 (TEM7) is included in the endothelial cells of tumors as a greatly expressed protein. In previous studies, it has been confirmed that TEM7 is highly expressed in gastric cancer (GC) cells and related to tumor invasion and migration. However, the relationship between TEM7 gene expressions, prognostic and tumor-infiltrating lymphocyte in GC is still unclear. Methods We obtained the expression of TEM7 in GC tissues using Oncomine and TIMER databases and validated it using qRT-PCR. The effect of TEM7 on survival in patients with GC was explored through the Kaplan–Meier Plotter database. Univariate and multifactorial analyses of the TCGA (The Cancer Genome Atlas) database were performed to explore the association of TEM7 expression with clinical traits. TIMER and GEPIA databases were used to analyze the relationship between TEM7 expression and immune infiltration. GSEA pathway enrichment in Kyoto Encyclopedia of Genes and Genomes (KEGG) exposed possible ways. Results An expression level of TEM7 was significantly increased in gastric cancer tissues and related with unfavorable survival in GC. Univariate analysis of TCGA (The Cancer Genome Atlas) database indicated that excessive level of TEM7 expression was linked with age, tumor grade and TMN classification, and multivariate analysis indicated that age and level of TEM7 expression were independent prognostic factors for a poor prognosis. The level of expression of TEM7 was positively related to tumor-infiltrating CD4+ and CD8+ T cells, neutrophils, macrophages, and dendritic cells other than B cells in GC. In the end, GSEA pathway enrichment exposed possible ways related to immunity. Discussion Our study indicates that TEM7 is a prognostic biomarker that makes the decision that the progression of GC and is connected with tumor immune infiltrates in GC.
Collapse
Affiliation(s)
- Lixin Geng
- Dalian Medical University, Dalian, Liaoning, 116044, People’s Republic of China
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Shuai Chen
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Yu Gong
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Yan Zhou
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Haojun Yang
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
| | - Liming Tang
- Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, People’s Republic of China
- Correspondence: Liming Tang Center of Gastrointestinal Disease, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, 68 Gehu Road, Wujin District, Changzhou, Jiangsu, People’s Republic of China Email
| |
Collapse
|
28
|
Zhong A, Chen T, Zhou T, Zhang Z, Shi M. TPD52L2 Is a Prognostic Biomarker and Correlated With Immune Infiltration in Lung Adenocarcinoma. Front Pharmacol 2021; 12:728420. [PMID: 34744715 PMCID: PMC8567320 DOI: 10.3389/fphar.2021.728420] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/20/2021] [Indexed: 01/12/2023] Open
Abstract
Tumor protein D52-like 2 (TPD52L2) belongs to the members of the TPD52 family. TPD52L2 was reported to regulate proliferation and apoptosis in cancer cells. However, its role in lung adenocarcinoma (LUAD) was uncertain. We evaluated the expression, methylation, copy number alteration, and prognostic significance of TPD52L2 using RNA-seq data from The Cancer Genome Atlas (TCGA). Enrichment analysis of TPD52L2 was conducted using the R package “clusterProfiler.” We further assessed the association between TPD52L2 and immune cell infiltration level, immunosuppressive genes, and tumor mutational burden (TMB). The difference of gene mutant frequency in high- and low-TPD52L2 groups was also analyzed. The results showed that TPD52L2 was over-expressed and predicted worse survival status in LUAD. We also found that TPD52L2 expression was positively associated with the infiltration levels of immunosuppressive cells, such as regulatory T cells (Tregs) and tumor-associated macrophages (TAMs), and negatively correlated with immune killer cells, such as CD8+ T and NK cells in pan-cancer, including LUAD. In addition, TPD52L2 expression was associated with immunosuppressive genes and TMB. High expression of TPD52L2 was with more mutant frequency of TP53. In summary, our results show that TPD52L2 is an oncogene and a potential prognostic biomarker in LUAD. High TPD52L2 expression is a possible indicator of immune infiltration and associated with tumor immunosuppressive status in LUAD.
Collapse
Affiliation(s)
- Anyuan Zhong
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ting Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Zhou
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zengli Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Minhua Shi
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Huang X, Zhang W, Pu F, Zhang Z. LncRNA MEG3 promotes chemosensitivity of osteosarcoma by regulating antitumor immunity via miR-21-5p/p53 pathway and autophagy. Genes Dis 2021; 10:531-541. [DOI: 10.1016/j.gendis.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 10/12/2021] [Accepted: 11/07/2021] [Indexed: 12/01/2022] Open
|
30
|
He J, Gao R, Meng M, Yu M, Liu C, Li J, Song Y, Wang H. Lysophosphatidic Acid Receptor 6 (LPAR6) Is a Potential Biomarker Associated with Lung Adenocarcinoma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182111038. [PMID: 34769557 PMCID: PMC8583018 DOI: 10.3390/ijerph182111038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 09/02/2021] [Indexed: 11/16/2022]
Abstract
LPAR6 is the most recently determined G-protein-coupled receptor of the lysophosphatidic acid receptor, and very few of studies have demonstrated the performance of LPAR6 in cancers. Moreover, the relationship of LPAR6 to the potential of prognosis and tumor infiltration immune cells in different types of cancer are still unclarified. In this study, the mRNA expression of LPAR6 and its clinical characteristics were evaluated on various databases. The association between LPAR6 and immune infiltrates of various types of cancer were investigated via TIMER. Immunohistochemistry (IHC) for LPAR6 in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) tissue microarray with patients’ information was detected. We constructed a systematic prognostic landscape in a variety of types of cancer base on the expression level of mRNA. We enclosed that higher LPAR6 mRNA expression level was associated with better overall survival in some types of malignancy. Moreover, LPAR6 significantly affects the prognostic potential of various cancers in The Cancer Genome Atlas Program (TCGA), especially in lung cancer. Tissue microarrays of lung cancer patient cohorts demonstrated that a higher protein level of LPAR6 was correlated to better overall survival of LUAD rather than LUSC cohorts. Further research indicated that the underlying mechanism of this phenome might be the mRNA expression level of LPAR6 was positively associated to infiltrating statuses of devious immunocytes in LUAD rather than in LUSC, that is, LPAR6 expression potentially contributes to the activation and recruiting of T cells (CD8+ T, naive T, effector T cell) and NK cells and inactivates Tregs, decreases T cell exhaustion and regulates T-helper (Th) cells in LUAD. Our discovery implies that LPAR6 is associated with prognostic potential and immune-infiltrating levels in LUAD. These discoveries imply that LPAR6 could be a promising novel biomarker for indicating the prognosis potential of LUAD patients.
Collapse
Affiliation(s)
- Jian He
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.G.); (M.M.); (J.L.)
- Correspondence: (J.H.); (Y.S.); (H.W.)
| | - Rui Gao
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.G.); (M.M.); (J.L.)
| | - Mei Meng
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.G.); (M.M.); (J.L.)
| | - Miao Yu
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
| | - Chengrong Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China;
| | - Jingquan Li
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.G.); (M.M.); (J.L.)
| | - Yizhi Song
- CAS Key Laboratory of Bio-Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou 215163, China;
- Correspondence: (J.H.); (Y.S.); (H.W.)
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; (R.G.); (M.M.); (J.L.)
- Correspondence: (J.H.); (Y.S.); (H.W.)
| |
Collapse
|
31
|
Yang WJ, Shi L, Wang XM, Yang GW. Heparanase is a novel biomarker for immune infiltration and prognosis in breast cancer. Aging (Albany NY) 2021; 13:20836-20852. [PMID: 34461608 PMCID: PMC8436937 DOI: 10.18632/aging.203489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/29/2021] [Indexed: 04/28/2023]
Abstract
Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate, regulates a variety of biological processes that promote tumor progression. In this study, we analyzed the correlation between HPSE expression and prognosis in cancer patients, using multiple databases (Oncomine, TIMER, PrognoScan, GEPIA, Kaplan-Meier plotter, miner v4.1, DAVID). HPSE expression was significantly increased in bladder, breast, lung, and stomach cancer compared to matched normal tissues. The increased HPSE expression correlated with poor prognosis and increased immune infiltration levels of B cells, CD8+ and CD4+ T cells, macrophages, neutrophils and dendritic cells in bladder and breast cancer. In breast cancer, the high HPSE expression was associated with basal-like subtypes, younger age (0-40), advanced Scarff-Bloom-Richardson grade, Nottingham Prognostic Index and p53 mutation status. In addition, using a mouse model of breast cancer, our data showed that HPSE upregulated IL-10 expression and promoted macrophage M2 polarization and T cell exhaustion. Together, our data provide a novel immunological perspective on the mechanisms underlying breast cancer progression, and indicate that HPSE may serve as a biomarker for immune infiltration and prognosis in breast cancer.
Collapse
Affiliation(s)
- Wen-Jing Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Lin Shi
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Xiao-Min Wang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| | - Guo-Wang Yang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing 100010, China
| |
Collapse
|
32
|
Alexander JL, Ibraheim H, Sheth B, Little J, Khan MS, Richards C, Hunter N, Chauhan D, Ratnakumaran R, McHugh K, Pinato DJ, Nathan P, Choy J, Crusz SM, Furness A, Turajlic S, Pickering L, Larkin J, Teare JP, Papa S, Speight A, Sharma A, Powell N. Clinical outcomes of patients with corticosteroid refractory immune checkpoint inhibitor-induced enterocolitis treated with infliximab. J Immunother Cancer 2021; 9:e002742. [PMID: 34233964 PMCID: PMC8264884 DOI: 10.1136/jitc-2021-002742] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/28/2022] Open
Abstract
INTRODUCTION Immune checkpoint inhibitors (CPIs) have changed the treatment landscape for many cancers, but also cause severe inflammatory side effects including enterocolitis. CPI-induced enterocolitis is treated empirically with corticosteroids, and infliximab (IFX) is used in corticosteroid-refractory cases. However, robust outcome data for these patients are scarce. METHODS We conducted a multicenter (six cancer centers), cohort study of outcomes in patients treated with IFX for corticosteroid-refractory CPI-induced enterocolitis between 2007 and 2020. The primary outcome was corticosteroid-free clinical remission (CFCR) with Common Terminology Criteria for Adverse Events (CTCAE) grade 0 for diarrhea at 12 weeks after IFX initiation. We also assessed cancer outcomes at 1 year using RECIST V1.1 criteria. RESULTS 127 patients (73 male; median age 59 years) were treated with IFX for corticosteroid-refractory CPI-induced enterocolitis. Ninety-six (75.6%) patients had diarrhea CTCAE grade >2 and 115 (90.6%) required hospitalization for colitis. CFCR was 41.2% at 12 weeks and 50.9% at 26 weeks. In multivariable logistic regression, IFX-resistant enterocolitis was associated with rectal bleeding (OR 0.19; 95% CI 0.04 to 0.80; p=0.03) and absence of colonic crypt abscesses (OR 2.16; 95% CI 1.13 to 8.05; p=0.03). Cancer non-progression was significantly more common in patients with IFX-resistant enterocolitis (64.4%) as compared with patients with IFX-responsive enterocolitis (37.5%; p=0.013). CONCLUSION This is the largest study to date reporting outcomes of IFX therapy in patients with corticosteroid-refractory CPI-induced enterocolitis. Using predefined robust endpoints, we have demonstrated that fewer than half of patients achieved CFCR. Our data also indicate that cancer outcomes may be better in patients developing prolonged and severe inflammatory side effects of CPI therapy.
Collapse
Affiliation(s)
- James L Alexander
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Hajir Ibraheim
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Bhavisha Sheth
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Jessica Little
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Muhammad Saheb Khan
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Camellia Richards
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Nikki Hunter
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Dharmisha Chauhan
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | | | - Kathleen McHugh
- Department of Gastroenterology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - David J Pinato
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, UK
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Paul Nathan
- Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Julia Choy
- Medical Oncology, Barts Health NHS Trust, London, UK
| | | | - Andrew Furness
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajlic
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
- Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, London, UK
| | - Lisa Pickering
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - James Larkin
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| | - Julian P Teare
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Sophie Papa
- Medical Oncology, Guy's and Saint Thomas' NHS Foundation Trust, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Ally Speight
- Department of Gastroenterology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Anand Sharma
- Medical Oncology, Mount Vernon Cancer Centre, Northwood, UK
| | - Nick Powell
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Oncology, Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
33
|
Liu Y, Xie S, Zhu K, Guan X, Guo L, Lu R. CALD1 is a prognostic biomarker and correlated with immune infiltrates in gastric cancers. Heliyon 2021; 7:e07257. [PMID: 34189308 PMCID: PMC8219766 DOI: 10.1016/j.heliyon.2021.e07257] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/17/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Background Caldesmon gene (CALD1) plays an important role in many cellular functions. Some researchers have found the correlation between CALD1 expression and prognosis of gastrointestinal cancer (GI), but the association with tumor-infiltrating lymphocytes (TILs) still unclear. Methods The expression of CALD1 in different human tumor was analyzed by Oncomine and Tumor Immune Estimation Resource (TIMER) databases. The correlations between CALD1 and prognosis in types cancer were explored by Kaplan–Meier plotter and Gene Expression Profiling Interactive Analysis (GEPIA) databases. The association between CALD1 expression and tumor immune cell infiltration was further analyzed via TIMER and GEPIA databases. Results The CALD1 expressions in types cancer between tumor tissues and adjacent normal tissues were significantly different. The high expression of CALD1 was related with poor overall survival (OS) of patients with gastric cancer, especially in gastric cancer patients at N1, N2 and N3 stages. The expression of CALD1 was positively associated with immune-infiltrated, such as CD8+T cells, CD4+T cells, macrophages, neutrophils, and dendritic cells (DCs) in gastric cancer. Conclusions CALD1 was considerably a key role in prognosis of patients with gastric cancer. The expression level of CALD1 is significantly associated with immune-infiltrated in gastric cancer. Furthermore, CALD1 expression may be involved in regulating tumor-associated macrophages (TAMs), dendritic cells, exhausted T cells and regulatory T cells in gastric cancer. These findings suggest that CALD1 could be utilized as a marker of prognosis and immune infiltration in gastric cancer. CALD1 plays an important role in immune infiltration in gastric cancer. CALD1 can affect the prognosis in gastric cancer patients with lymph node metastasis. CALD1 plays a vital role in immune escape in the gastric cancer microenvironment.
Collapse
Affiliation(s)
- Yixuan Liu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Suhong Xie
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China
| | - Keyu Zhu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaolin Guan
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Guo
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Renquan Lu
- Department of Clinical Laboratory, Fudan University Shanghai Cancer Center, No. 270, Dong'an Road, Xuhui District, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Nguyen H, Shah K, Waguespack SG, Hu MI, Habra MA, Cabanillas ME, Busaidy NL, Bassett R, Zhou S, Iyer PC, Simmons G, Kaya D, Pitteloud M, Subudhi SK, Diab A, Dadu R. Immune checkpoint inhibitor related hypophysitis: diagnostic criteria and recovery patterns. Endocr Relat Cancer 2021; 28:419-431. [PMID: 33890870 PMCID: PMC8183642 DOI: 10.1530/erc-20-0513] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/23/2021] [Indexed: 01/11/2023]
Abstract
Data on the diagnosis, natural course and management of immune checkpoint inhibitor (ICI)-related hypophysitis (irH) are limited. We propose this study to validate the diagnostic criteria, describe characteristics and hormonal recovery and investigate factors associated with the occurrence and recovery of irH. A retrospective study including patients with suspected irH at the University of Texas MD Anderson Cancer Center from 5/2003 to 8/2017 was conducted. IrH was defined as: (1) ACTH or TSH deficiency plus MRI changes or (2) ACTH and TSH deficiencies plus headache/fatigue in the absence of MRI findings. We found that of 83 patients followed for a median of 1.75 years (range 0.6-3), the proposed criteria used at initial evaluation accurately identified 61/62 (98%) irH cases. In the irH group (n = 62), the most common presentation was headache (60%), fatigue (66%), central hypothyroidism (94%), central adrenal insufficiency (69%) and MRI changes (77%). Compared with non-ipilimumab (ipi) regimens, ipi has a stronger association with irH occurrence (P = 0.004) and a shorter time to irH development (P < 0.01). Thyroid, gonadal and adrenal axis recovery occurred in 24, 58 and 0% patients, respectively. High-dose steroids (HDS) or ICI discontinuation was not associated with hormonal recovery. In the non-irH group (n = 19), one patient had isolated central hypothyroidism and six had isolated central adrenal insufficiency. All remained on hormone therapy at the last follow-up. We propose a strict definition of irH that identifies the vast majority of patients. HDS and ICI discontinuation is not always beneficial. Long-term follow-up to assess recovery is needed.
Collapse
Affiliation(s)
- Ha Nguyen
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Komal Shah
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Steven G Waguespack
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Mimi I Hu
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Mouhammed Amir Habra
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Maria E Cabanillas
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Naifa L Busaidy
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Roland Bassett
- Division of Science, Department of Biostatistics, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Shouhao Zhou
- Division of Science, Department of Biostatistics, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Priyanka C Iyer
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Garrett Simmons
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Diana Kaya
- Division of Diagnostic Imaging, Department of Diagnostic Radiology, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Marie Pitteloud
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Sumit K Subudhi
- Division of Cancer Medicine, Department of Genitourinary Medical Oncology, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Division of Cancer Medicine, Department of Melanoma Medical Oncology, The University of Texas Anderson Cancer Center, Houston, Texas, USA
| | - Ramona Dadu
- Division of Internal Medicine, Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas Anderson Cancer Center, Houston, Texas, USA
- Correspondence should be addressed to R Dadu;
| |
Collapse
|
35
|
Manz SM, Losa M, Fritsch R, Scharl M. Efficacy and side effects of immune checkpoint inhibitors in the treatment of colorectal cancer. Therap Adv Gastroenterol 2021; 14:17562848211002018. [PMID: 33948110 PMCID: PMC8053828 DOI: 10.1177/17562848211002018] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancers (CRCs) remain one of the most common and challenging neoplasia in the Western world. The response rate of immunotherapeutic treatment approaches in a subset of advanced CRCs is remarkable and has sustainably changed treatment regimens. Unfortunately, currently available immunotherapeutics only displayed significant antitumoral activity - in terms of progression free survival (PFS) and objective response rate (ORR) - in microsatellite instability-high (MSI-H)/DNA mismatch repair deficient (dMMR) CRCs. Subsequently, these remarkable results had led to the US Food and Drug Administration's approval of both immune checkpoint inhibitors (ICIs) pembrolizumab and nivolumab in the treatment of advanced MSI-H/dMMR CRCs. However, in microsatellite stable (MSS)/DNA mismatch repair proficient (pMMR) CRCs, ICIs have clearly failed to meet their expectations and are therefore not considered effective. As the vast majority of CRCs display a molecular MSS/pMMR profile, current treatment approaches endeavor to improve tumor immunogenicity that consecutively leads to increased proinflammatory cytokine levels as well as tumor infiltrating T-cells, which in turn may be targeted by various immunotherapeutic agents. Therefore, ongoing studies are investigating novel synergistic therapy modalities and approaches to overcome a "cold" to "hot" tumor conversion in MSS/pMMR CRCs. In this review, we summarize the efficacy and possible immune-related adverse events as well as novel therapeutic approaches of ICIs in the treatment of MSI-H/dMMR and MSS/pMMR CRCs.
Collapse
Affiliation(s)
- Salomon M. Manz
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marco Losa
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ralph Fritsch
- Center for Medical Oncology and Hematology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| |
Collapse
|
36
|
Qu S, Liu J, Wang H. EVA1B to Evaluate the Tumor Immune Microenvironment and Clinical Prognosis in Glioma. Front Immunol 2021; 12:648416. [PMID: 33889156 PMCID: PMC8056259 DOI: 10.3389/fimmu.2021.648416] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/15/2021] [Indexed: 12/16/2022] Open
Abstract
Background Previous research indicated that the tumor cells and microenvironment interactions are critical for the immunotherapeutic response. However, predicting the clinical response to immunotherapy remains a dilemma for clinicians. Hence, this study aimed to investigate the associations between EVA1B expression and prognosis and tumor-infiltrating immune cells in glioma. Methods Firstly, we detected the EVA1B expression in glioma tissues through biological databases. The chi-squared test, Kaplan-Meier, and univariate and multivariate Cox regression analyses were used to analyze the clinical significance of EVA1B expression. The correlation between EVA1B expression and levels of tumor-infiltrating immune cells in glioma tissues was investigated. Receiver operating characteristic (ROC) analysis was performed to compare the predictive power between EVA1B and other commonly immune-related markers. Results In the CGGA cohort of 325 glioma patients, we found that EVA1B was upregulated in glioma, and increased with tumor grade. High EVA1B expression was prominently associated with unfavorable clinicopathological features, and poorer survival of patients, which were further confirmed by TCGA (n=609) and GEO (n=74) cohorts. Furthermore, multivariate analysis indicated that EVA1B is an independent prognostic biomarker for glioma. Importantly, EVA1B overexpression was associated with a higher infiltration level of CD4+ T cells, CD8+ T cells, B cells, macrophages, and neutrophils in glioma. ROC curves showed that, compared with PD-L1, CTLA-4, and Siglec15, EVA1B presented a higher area under the curve (AUC) value (AUC=0.824) for predicting high immune infiltration levels in glioma. Conclusions We found that EVA1B was upregulated and could act as a poor prognostic biomarker in glioma. Importantly, EVA1B overexpression was associated with the immune infiltration levels of immune cells including B cells, CD4+ T cells, CD8+ T cells, macrophages, and neutrophils, and strongly with the overall immune infiltration levels of glioma. These findings suggested that EVA1B might be a potential biomarker for evaluating prognosis and immune infiltration in glioma.
Collapse
Affiliation(s)
- Shanqiang Qu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Liu
- Department of Neurosurgery, Lishui People's Hospital (The Sixth Affiliated Hospital of Wenzhou Medical University), Lishui, China
| | - Huafu Wang
- Department of Clinical Pharmacy, Lishui People's Hospital (The Sixth Affiliated Hospital of Wenzhou Medical University), Lishui, China
| |
Collapse
|
37
|
Goyal I, Pandey MR, Sharma R, Chaudhuri A, Dandona P. The side effects of immune checkpoint inhibitor therapy on the endocrine system. Indian J Med Res 2021; 154:559-570. [PMID: 35435341 PMCID: PMC9205006 DOI: 10.4103/ijmr.ijmr_313_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a relatively newer class of drugs approved for the treatment of malignancies such as melanoma, renal, bladder and lung cancer. Immune-related adverse events (IrAEs) involving the endocrine system are a common side effect of these drugs. The spectrum of endocrine adverse events varies by the drug class. Cytotoxic T-lymphocyte–associated antigen-4 inhibitors commonly cause hypophysitis/hypopituitarism, whereas the incidence of thyroid disease is higher with programmed cell death (PD)-1/ ligand (PD-L) protein 1 inhibitors. The focus of this review is to describe the individual endocrinopathies with their possible mechanisms, signs and symptoms, clinical assessment and disease management. Multiple mechanisms of IrAEs have been described in literature including type II/IV hypersensitivity reactions and development of autoantibodies. Patients with pre-existing autoimmune endocrine diseases can have disease exacerbation following ICI therapy rather than de novo IrAEs. Most of the endocrinopathies are relatively mild, and timely hormone replacement therapy allows continuation of ICIs. However, involvement of the pituitary–adrenal axis could be life-threatening if not recognized. Corticosteroids are helpful when the pituitary–adrenal axis is involved. In cases of severe endocrine toxicity (grade 3/4), ICIs should be temporarily discontinued and can be restarted after adequate hormonal therapy. Endocrinologists and general internists need to be vigilant and maintain a high degree of awareness for these adverse events.
Collapse
Affiliation(s)
- Itivrita Goyal
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| | - Manu Raj Pandey
- Department of Hematology & Oncology, State University of New York at Buffalo; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Rajeev Sharma
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo; Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ajay Chaudhuri
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| | - Paresh Dandona
- Department of Endocrinology, Diabetes & Metabolism, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
38
|
High Expression of PD-L1 Is Associated with Better Survival in Pancreatic/Periampullary Cancers and Correlates with Epithelial to Mesenchymal Transition. Diagnostics (Basel) 2021; 11:diagnostics11040597. [PMID: 33810560 PMCID: PMC8065840 DOI: 10.3390/diagnostics11040597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 01/02/2023] Open
Abstract
Periampullary cancers (PACs) are characterized by tumor-infiltrating lymphocytes (TILs), severe fibrosis, and epithelial to mesenchymal transition (EMT). The immune checkpoint marker programmed death-1 (PD-1) and its ligands 1 and 2 have gained popularity in cancers with TILs. Evidence suggests a strong relationship between immune checkpoint markers and EMT in cancers. Here, we evaluated the expression and prognostic significance of immune checkpoint and EMT markers in PAC using an automated image analyzer. Formalin-fixed, paraffin-embedded surgically excised PAC tissues from laboratory archives (1998–2014) were evaluated by immunohistochemical staining for PD-1, PD-L1, and PD-L2 in a tissue microarray. In total, 115 PAC patients (70 males and 45 females) with an average age of 63 years were analyzed. Location, gross type, size, radial resection margin, N-M stage, lymphatic invasion, vascular invasion, perineural invasion, histologically well-differentiated severe inflammation, and high PD-L1 expression were significantly associated with recurrence. Higher PD-L1 expression, but not PD-1 and PD-L2, was significantly related to better overall survival (OS) and disease-free survival (DFS). PD-L1 and PD-L2 were significantly related to EMT markers. Aside from other clinicopathologic parameters, high PD-L1 expression was significantly related to better OS and DFS of PAC patients. Moreover, immune checkpoint markers were significantly associated with EMT markers. Therefore, PD-L1 expression can be a good prognostic marker to guide future immune target-based therapies in PAC patients.
Collapse
|
39
|
Shah AA, Kamal MA, Akhtar S. Tumor Angiogenesis and VEGFR-2: Mechanism, Pathways and Current Biological Therapeutic Interventions. Curr Drug Metab 2021; 22:50-59. [PMID: 33076807 DOI: 10.2174/1389200221666201019143252] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 06/09/2020] [Accepted: 08/04/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Angiogenesis, involving the formation of new blood vessels from preexisting vessels, caters an important biological phenomenon for the growth and development of bodily structures in the human body. Regulation of angiogenesis in non-pathological conditions takes place through a well-defined balanced angiogenic-switch, which upon exposure to various pathological conditions may get altered. This makes the cells change their normal behavior resulting in uncontrolled division and angiogenesis. METHODS The current review tries to present a brief framework of angiogenesis and tumor progression phenomenon along with the latest therapeutic interventions against VEGFR-2 and its future directions. RESULTS The tumor angiogenic pathways functioning in diverse mechanisms via sprouting angiogenesis, intussusceptive angiogenesis, vascular co-option, vascular mimicry, and glomeruloid angiogenesis are normally activated by varied angiogenic stimulators and their receptors are interrelated to give rise to specialized signaling pathways. Amongst these receptors, VEGFR-2 is found as one of the key, critical mediators in tumor angiogenesis and is seen as a major therapeutic target for combating angiogenesis. Though a number of anti-angiogenic drugs like Ramucirumab, Sunitinib, Axitinib, Sorafenib, etc. showing good survival rates have been developed and approved by FDA against VEGFR-2, but these have also been found to be associated with serious health effects and adverse reactions. CONCLUSION An improved or alternative treatment is needed shortly that has a higher survival rate with the least side effects. Innovative strategies, including personalized medicine, nano-medicine, and cancer immunotherapy have also been outlined as an alternative treatment with a discussion on advancements and improvements required for their implementation methods.
Collapse
Affiliation(s)
- Altaf A Shah
- Department of Biosciences, Integral University, Lucknow-226026, UP, India
| | - Mohammad A Kamal
- Novel Global Community Educational Foundation, Peterlee Place, Hebersham, NSW 2770, Australia
| | - Salman Akhtar
- Novel Global Community Educational Foundation, Peterlee Place, Hebersham, NSW 2770, Australia
| |
Collapse
|
40
|
Kang Y, Niu Z, Hadlock T, Purcell E, Lo T, Zeinali M, Owen S, Keshamouni VG, Reddy R, Ramnath N, Nagrath S. On-Chip Biogenesis of Circulating NK Cell-Derived Exosomes in Non-Small Cell Lung Cancer Exhibits Antitumoral Activity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003747. [PMID: 33747745 PMCID: PMC7967048 DOI: 10.1002/advs.202003747] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/14/2020] [Indexed: 05/19/2023]
Abstract
As the recognition between natural killer (NK) cells and cancer cells does not require antigen presentation, NK cells are being actively studied for use in adoptive cell therapies in the rapidly evolving armamentarium of cancer immunotherapy. In addition to utilizing NK cells, recent studies have shown that exosomes derived from NK cells also exhibit antitumor properties. Furthermore, these NK cell-derived exosomes exhibit higher stability, greater modification potentials and less immunogenicity compared to NK cells. Therefore, technologies that allow highly sensitive and specific isolation of NK cells and NK cell-derived exosomes can enable personalized NK-mediated cancer therapeutics in the future. Here, a novel microfluidic system to collect patient-specific NK cells and on-chip biogenesis of NK-exosomes is proposed. In a small cohort of non-small cell lung cancer (NSCLC) patients, both NK cells and circulating tumor cells (CTCs) were isolated, and it is found NSCLC patients have high numbers of NK and NK-exosomes compared with healthy donors, and these concentrations show a trend of positive and negative correlations with bloodborne CTC numbers, respectively. It is further demonstrated that the NK-exosomes harvested from NK-graphene oxide chip exhibit cytotoxic effect on CTCs. This versatile system is expected to be used for patient-specific NK-based immunotherapies along with CTCs for potential prognostic/diagnostic applications.
Collapse
Affiliation(s)
- Yoon‐Tae Kang
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Zeqi Niu
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Thomas Hadlock
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Emma Purcell
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Ting‐Wen Lo
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Mina Zeinali
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | - Sarah Owen
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
| | | | - Rishindra Reddy
- Michigan Medicine Thoracic Surgery ClinicTaubman Center1500E Medical Center Dr. SPC 5344Ann ArborMI48109USA
| | - Nithya Ramnath
- Department of Internal MedicineUniversity of MichiganAnn ArborMI48109USA
| | - Sunitha Nagrath
- Department of Chemical EngineeringBiointerfaces InstituteUniversity of MichiganAnn ArborMI48109USA
- Rogel Cancer CenterUniversity of Michigan1500 East Medical Center DriveAnn ArborMI48109USA
| |
Collapse
|
41
|
Wang Y, Mohseni M, Grauel A, Diez JE, Guan W, Liang S, Choi JE, Pu M, Chen D, Laszewski T, Schwartz S, Gu J, Mansur L, Burks T, Brodeur L, Velazquez R, Kovats S, Pant B, Buruzula G, Deng E, Chen JT, Sari-Sarraf F, Dornelas C, Varadarajan M, Yu H, Liu C, Lim J, Hao HX, Jiang X, Malamas A, LaMarche MJ, Geyer FC, McLaughlin M, Costa C, Wagner J, Ruddy D, Jayaraman P, Kirkpatrick ND, Zhang P, Iartchouk O, Aardalen K, Cremasco V, Dranoff G, Engelman JA, Silver S, Wang H, Hastings WD, Goldoni S. SHP2 blockade enhances anti-tumor immunity via tumor cell intrinsic and extrinsic mechanisms. Sci Rep 2021; 11:1399. [PMID: 33446805 PMCID: PMC7809281 DOI: 10.1038/s41598-021-80999-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
SHP2 is a ubiquitous tyrosine phosphatase involved in regulating both tumor and immune cell signaling. In this study, we discovered a novel immune modulatory function of SHP2. Targeting this protein with allosteric SHP2 inhibitors promoted anti-tumor immunity, including enhancing T cell cytotoxic function and immune-mediated tumor regression. Knockout of SHP2 using CRISPR/Cas9 gene editing showed that targeting SHP2 in cancer cells contributes to this immune response. Inhibition of SHP2 activity augmented tumor intrinsic IFNγ signaling resulting in enhanced chemoattractant cytokine release and cytotoxic T cell recruitment, as well as increased expression of MHC Class I and PD-L1 on the cancer cell surface. Furthermore, SHP2 inhibition diminished the differentiation and inhibitory function of immune suppressive myeloid cells in the tumor microenvironment. SHP2 inhibition enhanced responses to anti-PD-1 blockade in syngeneic mouse models. Overall, our study reveals novel functions of SHP2 in tumor immunity and proposes that targeting SHP2 is a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Wang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Morvarid Mohseni
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Angelo Grauel
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Javier Estrada Diez
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Wei Guan
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Simon Liang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jiyoung Elizabeth Choi
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Minying Pu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Dongshu Chen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Tyler Laszewski
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Stephanie Schwartz
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jane Gu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Leandra Mansur
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Tyler Burks
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Lauren Brodeur
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Roberto Velazquez
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Steve Kovats
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Bhavesh Pant
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Giri Buruzula
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Emily Deng
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Julie T Chen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Farid Sari-Sarraf
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Christina Dornelas
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Malini Varadarajan
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Haiyan Yu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Chen Liu
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Joanne Lim
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Huai-Xiang Hao
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Xiaomo Jiang
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Anthony Malamas
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Matthew J LaMarche
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Felipe Correa Geyer
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Margaret McLaughlin
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Carlotta Costa
- Oncology Disease Area, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Joel Wagner
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - David Ruddy
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Pushpa Jayaraman
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | | | - Pu Zhang
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Oleg Iartchouk
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Cambridge, USA
| | - Kimberly Aardalen
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Viviana Cremasco
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Glenn Dranoff
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Jeffrey A Engelman
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Serena Silver
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Hongyun Wang
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - William D Hastings
- Exploratory Immuno-Oncology, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| | - Silvia Goldoni
- Oncology Disease Area, Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
42
|
Xiang A, Lin X, Xu L, Chen H, Guo J, Zhou F. PCOLCE Is Potent Prognostic Biomarker and Associates With Immune Infiltration in Gastric Cancer. Front Mol Biosci 2020; 7:544895. [PMID: 33392251 PMCID: PMC7775582 DOI: 10.3389/fmolb.2020.544895] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/08/2020] [Indexed: 12/25/2022] Open
Abstract
Background The exact biological role of PCOLCE was not yet clear and there were few reports study the correlation of PCOLCE gene expression level with the occurrence and development of gastric cancer. Methods The expression of PCOLCE was analyzed by performing the Oncomine and Ualcan database. We evaluated the function of PCOLCE on clinical prognosis with the use of Kaplan-Meier plotter database. The relationship between PCOLCE and cancer immune in filtrates was researched by Tumor Immune Estimation Resource (TIMER) site database. Results PCOLCE significantly upregulated in gastric cancer patients compared to normal gastric samples. And the increased expression of PCOLCE mRNA was closely linked to shorter overall survival (OS), progress-free survival (PFS) in all gastric cancers. Besides, PCOLCE expression displayed a tight correlation with infiltrating levels of macrophages and dendritic cells (DCs) in gastric cancer. Moreover, PCOLCE expression was positively correlated with diverse immune marker sets in gastric cancer. Conclusion All the results above suggested that overexpression of PCOLCE indicated unfavorable prognosis in patients with gastric cancer. PCOLCE was correlated with immune infiltrating levels including those of B cells, CD8 + T cells, CD4 + T cells, macrophages, neutrophils, and DCs in gastric cancer patients. All the findings suggested that PCOLCE could be used as a prognostic biomarker for determining prognosis and immune infiltration in gastric cancer. Additionally, PCOLCE expression potentially contributed to the regulation of monocyte, M2 macrophage, Tfh, CD8 + T cell, TAM, Th1 cell Thus PCOLCE is a potential target for gastric cancer therapy and these preliminary findings require further study to determine whether PCOLCE-targeting reagents might be developed for clinical application in gastric cancer.
Collapse
Affiliation(s)
- Aizhai Xiang
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Lin
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lvping Xu
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Honggang Chen
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jufeng Guo
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Zhou
- Hangzhou First People's Hospital, Hangzhou, China.,Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
43
|
Tu Y, Han J, Dong Q, Chai R, Li N, Lu Q, Xiao Z, Guo Y, Wan Z, Xu Q. TGF-β2 is a Prognostic Biomarker Correlated with Immune Cell Infiltration in Colorectal Cancer: A STROBE-compliant article. Medicine (Baltimore) 2020; 99:e23024. [PMID: 33181667 PMCID: PMC7668458 DOI: 10.1097/md.0000000000023024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Transforming growth factor-beta (TGF-β2) is an important cytokine regulating immune cell function. However, whether TGF-β2 controls the invasion of colorectal cancer (CRC) by immune cells is unknown. Therefore, we evaluated the expression of TGF-β2 using multiple databases and determined the relationship between TGF-β2 expression and tumor immune infiltration defined by a set of genetic markers. The analysis demonstrated that the expression of TGF-β2 is closely related to the outcome of many cancers, and this correlation was particularly strong in CRC. In addition, the increased expression of TGF-β2 was significantly associated with the expression of various markers of specific immune cell subpopulations, and overexpression of TGF-β2 was closely related to the prognosis of colon cancer patients. Moreover, TGF-β2 was related to the prognosis and infiltration of the tumor by immune cells in CRC patients. The obtained results indicate that TGF-β2 is a critical factor regulating the recruitment of immune cells and controls their infiltration into colorectal tumors. Thus, high expression of TGF-β2 not only facilitates the prognosis in CRC patients, but also may provide a new target for the treatment of CRC.
Collapse
Affiliation(s)
- Yifeng Tu
- Department of Pathology, College of Basic Medical, Sciences, Shenyang Medical College, Shenyang, Liaoning
| | - Junjun Han
- Graduate Department, BengBu Medical College, Bengbu, Anhui
| | - Quanjin Dong
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medica College), Hangzhou, Zhejiang
| | - Rui Chai
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medica College), Hangzhou, Zhejiang
| | - Na Li
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's
| | - Qiliang Lu
- Medical Department, Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Zunqiang Xiao
- The Second Clinical Medical Department, Zhejiang Chinese Medical University
| | - Yang Guo
- Department of Colorectal Surgery, Zhejiang Provincial People's Hospital People's Hospital of Hangzhou Medical College), Hangzhou, Zhejiang
| | - Ziang Wan
- Zhejiang Provincial People's Hospital
| | - Qiuran Xu
- The Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's
| |
Collapse
|
44
|
Zhang Z, Zhang F, Yuan F, Li Y, Ma J, Ou Q, Liu Z, Yang B, Wang L, Tao H, Zhang S, Li X, Zhi X, Ge X, Bao H, Wu X, Hu Y, Wang J. Pretreatment hemoglobin level as a predictor to evaluate the efficacy of immune checkpoint inhibitors in patients with advanced non-small cell lung cancer. Ther Adv Med Oncol 2020; 12:1758835920970049. [PMID: 33224276 PMCID: PMC7649885 DOI: 10.1177/1758835920970049] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/02/2020] [Indexed: 02/06/2023] Open
Abstract
Background: Targeting immune checkpoints represents an immense breakthrough in cancer therapeutics. The prognostic value of hemoglobin (Hb) has been investigated in many malignancies including non-small cell lung cancer (NSCLC). However, the prognostic impact of pretreatment Hb count for immune checkpoint inhibitors (ICIs) in advanced NSCLC patients remains unclear. Methods: A total of 310 late-stage NSCLC patients who received ICI therapies between January 2015 and March 2019 were prospectively enrolled. We used a propensity score-matched cohort analysis for this study. Patients’ clinicopathological characteristics and pretreatment Hb concentration were assessed against the progression-free survival (PFS) and overall survival (OS) using the Kaplan–Meier method and Cox proportional hazards regression. Results: A propensity score (PS)-matched cohort analysis was applied to adjust for potential bias and to create two comparable groups according to patients’ clinicopathological characteristics. The patients with normal baseline Hb levels (⩾110 g/L) had significantly longer PFS [median: 10.0 versus 4.0 months, hazard ratio (HR): 0.63, 95% confidence interval (CI): 0.46−0.86; p = 0.001] and OS [median: 17.6 versus 10.5 months, HR (95% CI): 0.56 (0.40−0.79); p < 0.001] than those with decreased Hb count (<110 g/L) in a PS-matched cohort (n = 255). For patients with normal pretreatment Hb levels, ICI combination therapy was significantly associated with better PFS [median: 11.1 versus 8.0 months, HR (95% CI): 0.74 (0.50−1.06); p = 0.09] and OS [median: 26.0 versus 12.9 months, HR (95% CI): 0.56 (0.37−0.86); p = 0.008] than monotherapy, but there was no such trend for patients with decreased baseline Hb levels. Conclusion: Our findings showed that normal pretreatment Hb count served as a favorable prognostic marker in advanced NSCLC patients treated with ICIs, representing an economical biomarker with readily measuring performance among all reported ones.
Collapse
Affiliation(s)
- Zhibo Zhang
- Department of Oncology, The Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fan Zhang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Fang Yuan
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Ye Li
- Department of Radiotherapy, Chinese PLA General Hospital, Beijing, China
| | - Junxun Ma
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Qiuxiang Ou
- Translational Medicine Research Institute, Geneseeq Technology Inc., Toronto, Ontario, Canada
| | - Zhefeng Liu
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Bo Yang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Lijie Wang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Haitao Tao
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Sujie Zhang
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoyan Li
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Xiaoyu Zhi
- Department of Oncology, The Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiangwei Ge
- Department of Oncology, The Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hua Bao
- Translational Medicine Research Institute, Geneseeq Technology Inc., Toronto, Ontario, Canada
| | - Xue Wu
- Translational Medicine Research Institute, Geneseeq Technology Inc., Toronto, Ontario, Canada
| | - Yi Hu
- Department of Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China
| | - Jinliang Wang
- Department of Oncology, The Second Medical Center of Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing, China
| |
Collapse
|
45
|
Ibraheim H, Baillie S, Samaan MA, Abu-Sbeih H, Wang Y, Talley NJ, P Jones M, Powell N. Systematic review with meta-analysis: effectiveness of anti-inflammatory therapy in immune checkpoint inhibitor-induced enterocolitis. Aliment Pharmacol Ther 2020; 52:1432-1452. [PMID: 32920854 DOI: 10.1111/apt.15998] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 03/29/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors have revolutionised cancer treatment, but at the cost of off-target immune-mediated organ damage. This includes checkpoint inhibitor-induced enterocolitis which frequently requires hospitalisation and may be life-threatening. Empiric treatment typically includes corticosteroids and infliximab, although no large-scale studies have confirmed their effectiveness. AIM To investigate the effectiveness of anti-inflammatory therapy in checkpoint inhibitor-induced enterocolitis METHODS: We performed a systematic review and meta-analysis of studies reporting clinical outcomes of checkpoint inhibitor-induced enterocolitis in adult cancer patients treated with anti-inflammatory agents. We searched Medline, EMBASE, and the Cochrane library through April and extracted the proportion of patients responding to anti-inflammatory therapy. Variation in effect size was studied using a random-effects meta-regression analysis, with checkpoint inhibitor agent and tumour type as the variables. RESULTS Data were pooled from 1210 treated patients across 39 studies. Corticosteroids were effective in 59% (95% CI 54- 65) of patients, with response significantly more favourable in patients treated with anti-PD-1/L1 monotherapy, compared with anti-CTLA-4 containing regimens (78%, 95% CI 69-85 vs 56 %, 95% CI 49-63, P = 0.003), and more favourable in lung cancer patients compared with melanoma patients (88%, 95% CI 62-97 vs 55%, 95% CI 47-63, P = 0.04). Infliximab was effective in 81% (95% CI 73-87) of patients, and vedolizumab in 85% (95% CI 60-96). CONCLUSION Corticosteroids, infliximab and vedolizumab, are effective in the treatment of checkpoint inhibitor-induced enterocolitis. Checkpoint inhibitor regimen and cancer type were significant moderators in response to corticosteroid therapy.
Collapse
Affiliation(s)
- Hajir Ibraheim
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK.,The Royal Marsden Hospital, London, UK
| | | | - Mark A Samaan
- Department of Gastroenterology, Guy's and St Thomas' Hospital, London, UK
| | | | - Yinghong Wang
- The University of Texas MD Anderson Cancer Cente, Houston, TX, USA
| | | | - Michael P Jones
- Psychology Department, Macquarie University, North Ryde, NSW, Australia
| | - Nick Powell
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK.,The Royal Marsden Hospital, London, UK
| |
Collapse
|
46
|
Dang H, Sun J, Wang G, Renner G, Layfield L, Hilli J. Management of pembrolizumab-induced steroid refractory mucositis with infliximab: A case report. World J Clin Cases 2020; 8:4100-4108. [PMID: 33024767 PMCID: PMC7520797 DOI: 10.12998/wjcc.v8.i18.4100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/30/2020] [Accepted: 08/21/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Pembrolizumab is an anti-programmed death receptor 1 (PD-1) that was shown to have a tolerable safety profile with 17% of grade 3-4 drug-related adverse events, notable response rate of 16% with median duration of response of 8 mo, and median overall survival of 8 mo. Severe mucositis is a very rare complication with only two cases of grade 4 mucositis reported, and both cases had good response to intravenous methylprednisolone and subsequent oral prednisone tapering. We report the first case of pembrolizumab-induced severe mucositis that was refractory to steroid treatment.
CASE SUMMARY An 80-year-old woman with a past medical history of recurrent right cheek nodular melanoma status post resection and new right lung metastatic melanoma on immunotherapy presented with dysphagia and odynophagia for 2 mo. She initially received 2 doses of ipilimumab 1 year ago with good outcome, but treatment was discontinued after developing severe diarrhea and rash. Pembrolizumab was then initiated 4 mo after disease progression. Significant improvement was noted after 3 doses. However, after 6 cycles of pembrolizumab, patient developed odynophagia and malnutrition. Improvement of symptoms was noted after discontinuation of pembrolizumab and initiation of steroids. 3 mo later, patient developed pharyngeal swelling with hoarseness and new oxygen requirement due to impending airway obstruction while being on prednisone tapering regimen, finally ended up with intubation and tracheostomy. Histologic analysis of left laryngeal and epiglottis tissue showed granulation tissue with acute on chronic inflammation, negative for malignancy and infection. Patient achieved marked improvement after 2 doses of infliximab of 5 mg/kg every 2 wk while continuing on prednisone tapering course.
CONCLUSION We report the first case of pembrolizumab-induced grade 4 mucositis that had limited recovery with prolonged steroid course but had rapid response with addition of infliximab. The patient had recurrent mucositis symptoms whenever steroids was tapered but achieved complete response after receiving two doses of infliximab while continuing to be on tapering steroids. The success of infliximab in this patient with pembrolizumab-induced severe mucositis presents a potentially safe approach to reduce prolonged steroid course and accelerate recovery in managing this rare complication.
Collapse
Affiliation(s)
- Harry Dang
- Department of Medicine, University of Missouri, Columbia, MO 65212, United States
| | - Jiyuan Sun
- Department of Hematology-Oncology, University of Missouri, Columbia, MO 65212, United States
| | - Guoliang Wang
- Department of Pathology, University of Missouri, Columbia, MO 65212, United States
| | - Gregory Renner
- Department of Otolaryngology - Head and Neck Surgery, University of Missouri, Columbia, MO 65212, United States
| | - Lester Layfield
- Department of Pathology, University of Missouri, Columbia, MO 65212, United States
| | - Jaffar Hilli
- Department of Hematology-Oncology, University of Missouri, Columbia, MO 65212, United States
| |
Collapse
|
47
|
Chang M, Lally SE, Dalvin LA, Orloff MM, Shields CL. Conjunctival melanoma with orbital invasion and liver metastasis managed with systemic immune checkpoint inhibitor therapy. Indian J Ophthalmol 2020; 67:2071-2073. [PMID: 31755463 PMCID: PMC6896557 DOI: 10.4103/ijo.ijo_663_19] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
A 60-year-old Caucasian female was referred for biopsy-proven amelanotic orbito-conjunctival melanoma. Map biopsies revealed residual invasive melanoma on the deep tarsal margin at the site of previous surgery. Repeat excisions were required after recurrence was detected following 3 months and 7 months. Positron emission tomography scan detected liver metastasis and additional orbito-conjunctival melanoma recurrence. Biomarker testing showed NRAS mutation without BRAF or c-KIT mutations and without PD-L1 expression. Systemic checkpoint inhibitor therapy was initiated with regression of both the orbito-conjunctival melanoma and liver metastasis. Invasive, non resectable orbito-conjunctival melanoma with liver metastasis can demonstrate a response to systemic checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Michael Chang
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sara E Lally
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lauren A Dalvin
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA; Department of Ophthalmology, Mayo Clinic, Rochester, MN, USA
| | - Marlana M Orloff
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Carol L Shields
- Ocular Oncology Service, Wills Eye Hospital, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
48
|
SNAI1 is a prognostic biomarker and correlated with immune infiltrates in gastrointestinal cancers. Aging (Albany NY) 2020; 12:17167-17208. [PMID: 32833672 PMCID: PMC7521489 DOI: 10.18632/aging.103667] [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: 02/17/2020] [Accepted: 06/04/2020] [Indexed: 01/24/2023]
Abstract
Epithelial-mesenchymal transition (EMT)-related genes play an important role in immunosuppression. However, the correlations of EMT-related genes to prognosis and tumor-infiltrating lymphocytes in different cancers remain unclear. TCGA, GEO databases were used to analyze the expression, prognosis, and immune infiltration of EMT markers in cancer. RT-qPCR, immunohistochemistry, and western blot were used to analysis the expression and prognosis of SNAI1 in gastrointestinal cancers. High SNAI1 expression was closely related with poorer overall survival in gastrointestinal cancers in TCGA cohort. High SNAI1 expression was closely related with poorer overall survival in gastrointestinal cancers, and was validated in GEO database. Simultaneously, high expression of SNAI1 correlates with clinical relevance of gastric cancer. Moreover, SNAI1 expression was associated with tumor-infiltrating immune cells in gastrointestinal cancers. In addition, RT-qPCR, immunohistochemistry, and western blot showed SNAI1 expression was higher in gastrointestinal cancers compared to the normal tissues. Finally, high SNAI1 expression was closely related with poorer overall survival and correlates with clinical relevance of gastrointestinal cancers in an independent validation cohort. In summary, the results approaches to suggest that SNAI1 can be used as a prognostic biomarker for determining prognosis and immune infiltration in gastrointestinal cancers.
Collapse
|
49
|
D'Arrigo P, Tufano M, Rea A, Vigorito V, Novizio N, Russo S, Romano MF, Romano S. Manipulation of the Immune System for Cancer Defeat: A Focus on the T Cell Inhibitory Checkpoint Molecules. Curr Med Chem 2020; 27:2402-2448. [PMID: 30398102 DOI: 10.2174/0929867325666181106114421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 10/15/2018] [Accepted: 10/24/2018] [Indexed: 12/19/2022]
Abstract
The immune system actively counteracts the tumorigenesis process; a breakout of the immune system function, or its ability to recognize transformed cells, can favor cancer development. Cancer becomes able to escape from immune system control by using multiple mechanisms, which are only in part known at a cellular and molecular level. Among these mechanisms, in the last decade, the role played by the so-called "inhibitory immune checkpoints" is emerging as pivotal in preventing the tumor attack by the immune system. Physiologically, the inhibitory immune checkpoints work to maintain the self-tolerance and attenuate the tissue injury caused by pathogenic infections. Cancer cell exploits such immune-inhibitory molecules to contrast the immune intervention and induce tumor tolerance. Molecular agents that target these checkpoints represent the new frontier for cancer treatment. Despite the heterogeneity and multiplicity of molecular alterations among the tumors, the immune checkpoint targeted therapy has been shown to be helpful in selected and even histologically different types of cancer, and are currently being adopted against an increasing variety of tumors. The most frequently used is the moAb-based immunotherapy that targets the Programmed Cell Death 1 protein (PD-1), the PD-1 Ligand (PD-L1) or the cytotoxic T lymphocyte antigen-4 (CTLA4). However, new therapeutic approaches are currently in development, along with the discovery of new immune checkpoints exploited by the cancer cell. This article aims to review the inhibitory checkpoints, which are known up to now, along with the mechanisms of cancer immunoediting. An outline of the immune checkpoint targeting approaches, also including combined immunotherapies and the existing trials, is also provided. Notwithstanding the great efforts devoted by researchers in the field of biomarkers of response, to date, no validated FDA-approved immunological biomarkers exist for cancer patients. We highlight relevant studies on predictive biomarkers and attempt to discuss the challenges in this field, due to the complex and largely unknown dynamic mechanisms that drive the tumor immune tolerance.
Collapse
Affiliation(s)
- Paolo D'Arrigo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Martina Tufano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Anna Rea
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Vincenza Vigorito
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Nunzia Novizio
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Salvatore Russo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Maria Fiammetta Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| | - Simona Romano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
50
|
Lyu L, Zheng Y, Hong Y, Wang M, Deng Y, Wu Y, Xu P, Yang S, Wang S, Yao J, Zhang D, Guo Y, Lyu J, Dai Z. Comprehensive analysis of the prognostic value and immune function of chemokine-CXC receptor family members in breast cancer. Int Immunopharmacol 2020; 87:106797. [PMID: 32702599 DOI: 10.1016/j.intimp.2020.106797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/23/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022]
Abstract
Recently, immune checkpoint inhibitors (ICIs) have been successfully used for treating melanoma. Unfortunately, many breast cancer (BC) patients show low response to ICIs due to the lack of infiltrating immune cells. Previous studies revealed that chemokine-CXC receptors (CXCRs) play a crucial role in leukocyte infiltration and promote cancer cell proliferation, migration, metastasis, and angiogenesis. However, the underlying functions of CXCRs in cancer-immunity cycle remain unclear. In this study, we firstly found that in comparison to normal tissues, BC tissues, especially basal-like BC, showed increased mRNA levels of CXCR3/4/5/6/8, but decreased CXCR1/2/7 expression using UALCAN and TIMER database. Interestingly, it's was found that the mRNA levels of CXCR3/4/5/6 were decreased in lymphocyte depleted of the BC immune subtype. Subsequently, functional enrichment analysis of distinct CXCRs indicated that CXCR3/4/5/6 were strongly associated to immune-related biological functions. Therefore, further analysis using TIMER and TISIDB database suggested that CXCR3/4/5/6 expression were strongly correlated with tumor-infiltrating lymphocytes (TILs) and immune checkpoints in BC. Finally, Kaplan-Meier Plotter analysis indicated that high mRNA expression of CXCR4 predicted worse relapse-free survival (RFS), whereas CXCR3/5/6 indicated better RFS in BC patients. These findings suggest a therapeutic value for CXCR3/4/5/6 in combination with ICIs for the treatment of BC.
Collapse
Affiliation(s)
- Lijuan Lyu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yun Hong
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Meng Wang
- Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yujiao Deng
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Wu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Peng Xu
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Si Yang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuqian Wang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jia Yao
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Dai Zhang
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China; Department of Oncology, The 2nd Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Guo
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Jun Lyu
- Department of Clinical Research, The First Affiliated Hospital of Jinan University, Guangzhou, China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|