167 results on '"immune checkpoint blockade (ICB)"'
Search Results
2. UBR5 in Tumor Biology: Exploring Mechanisms of Immune Regulation and Possible Therapeutic Implications in MPNST.
- Author
-
Odhiambo, Diana Akinyi, Fan, Selina, and Hirbe, Angela C.
- Subjects
- *
IMMUNOGENETICS , *SURVIVAL rate , *NEOPLASTIC cell transformation , *IMMUNOTHERAPY , *NEUROFIBROMATOSIS 1 , *ENZYMES , *NERVOUS system tumors , *METASTASIS , *IMMUNE checkpoint inhibitors , *ONCOGENES , *SOFT tissue tumors , *IMMUNITY - Abstract
Simple Summary: Malignant peripheral nerve sheath tumor (MPNST) is an aggressive soft-tissue cancer with limited treatment options and poor survival rates, especially in cases with unresectable or metastatic disease. This review focuses on the role of UBR5, a gene frequently amplified in cancers and linked to immune suppression and tumor growth. Due to the typically low immune activity in MPNST, immune checkpoint blockade therapies—which have shown promise in treating metastatic cancers—are less effective. Studies in other cancers show that UBR5 can influence immune responses and tumor progression, indicating it might play a similar role in MPNST. By exploring UBR5's regulatory impact on the immune landscape, this review aims to provide insights into potential novel therapeutic strategies for inoperable and metastatic cases of MPNST. Malignant peripheral nerve sheath tumor (MPNST) is a rare but aggressive soft-tissue sarcoma characterized by poor response to therapy. The primary treatment remains surgical resection with negative margins. Nonetheless, in the setting of neurofibromatosis type 1 (NF1), the five-year survival rate is at 20–50%, with recurrence occurring in up to 50% of individuals. For patients with metastatic and unresectable disease, current treatment options include cytotoxic chemotherapy, which offers minimal benefit, and most patients die within five years of diagnosis. Despite advances in targeted therapy focusing on inhibiting Ras signaling and its downstream effectors, clinical trials report minimal clinical benefit, highlighting the need to explore alternative pathways in MPNST pathogenesis. Here, we discuss the role of the E3 ubiquitin ligase, UBR5, in cancer progression and immune modulation across various malignancies, including breast, lung, and ovarian cancer. We focus on mechanisms by which UBR5 contributes to tumorigenesis, focusing on its influence on tumor microenvironment and immune modulation. Additionally, we explore UBR5's roles in normal tissue function, DNA damage response, metastasis, and therapeutic resistance, illustrating its multifaceted contribution to cancer biology. We discuss evidence implicating UBR5 in immune evasion and highlight its potential as a therapeutic target to enhance the efficacy of immune checkpoint blockade (ICB) therapy in MPNST, a tumor typically characterized by an immune cold microenvironment. We outline current immune-based strategies and challenges in MPNST management, ongoing efforts to shift the immune landscape in MPNST, and ultimately, we suggest that targeting UBR5 could be a novel strategy to potentiate ICB therapy-mediated anti-tumor immune response and clinical outcomes, particularly in MPNST patients with inoperable or metastatic disease. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
3. Type I Diabetes Mellitus impairs cytotoxic immunity through CEACAM5 upregulation in colorectal cancer: Exploring the intersection of autoimmune dysfunction and cancer progression: the role of NF-κB p65 in colorectal cancer.
- Author
-
Yingying, Li, Xingyong, Feng, Deying, Zhao, Xingchen, Tian, Jiahua, Zou, and Jie, Yu
- Abstract
Type 1 diabetes (T1D) is characterized by an autoimmune-mediated destruction of pancreatic beta cells and a chronic inflammatory state, which may influence the progression of colorectal cancer (CRC) through immune system dysregulation and enhanced tumor immune evasion. This study aims to elucidate the role of p65 in modulating the tumor microenvironment in CRC within the context of T1D and to determine how this modulation affects tumor growth, immune cell infiltration, and the expression of immune evasion molecules such as CEACAM5. NOD mice, which model T1D, were inoculated with MC38 colon carcinoma cells engineered to knock down p65. Tumor growth was monitored, and the tumor microenvironment was analyzed using flow cytometry to assess the infiltration of immune cells. The expression of Ki-67 and CEACAM5 in tumor cells was also evaluated. Additionally, in vitro assays were conducted to study the proliferation and activation of T cells co-cultured with tumor cells. Knockdown of p65 in tumor cells significantly inhibited tumor growth in NOD mice. This was accompanied by an increased infiltration of cytotoxic CD8+ T cells and no significant change in CD4+ or Foxp3 + T regulatory cells within the tumor microenvironment. There was a notable reduction in the expression of Ki-67 and CEACAM5, indicating decreased proliferation and potential immune evasion capabilities of the tumor cells. Our findings demonstrate that the NF-κB p65 subunit plays a crucial role in promoting tumor growth and modulating the immune microenvironment in CRC, particularly in the context of T1D. Knocking down p65 not only reduces tumor progression but also enhances the anti-tumor immune response by decreasing immune evasion mechanisms. These results suggest that targeting the NF-κB pathway may be a viable strategy to improve the efficacy of cancer immunotherapy, especially in patients with autoimmune diseases like T1D. Physical activity enhances the effect of immune checkpoint blockade by inhibiting the intratumoral HIF1-α/CEACAM5 axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Characterisation of the tumour immune microenvironment and mechanisms of checkpoint blockade in mesothelioma
- Author
-
Harber, James
- Subjects
Malignant pleural mesothelioma (MPM) ,Cancer ,Mesothelioma ,Tumour Immune Microenvironment ,Immune checkpoint blockade (ICB) ,Thesis - Abstract
Malignant pleural mesothelioma (MPM) is a lethal cancer causally associated with asbestos exposure. MPM carries a dismal prognosis and few effective treatment options currently exist. Immune checkpoint blockade (ICB) immunotherapies have repeatedly demonstrated efficacy in clinical trials for MPM since 2017 and have received regulatory approval in many industrialised countries since 2020. Despite its efficacy, the mechanisms behind these ICB responses remain unknown, partially because many canonical pan-cancer biomarkers are less reliable in MPM, but also due to a scarcity of translational data. This project therefore aimed to characterise MPM's complex tumour immune microenvironment and elucidate mechanisms of response and resistance to ICB, focussing on tumour-associated macrophages (TAMs) and cytotoxic T lymphocytes (CTLs). Bioinformatic deconvolution algorithms, a syngeneic mouse model and multiplex fluorescent immunohistochemistry were used to quantify these key leukocytes and evaluate their clinical impact. Deconvolution-defined TAMs were associated with poor clinical outcomes and lower CTL infiltration, though inter-algorithm discrepancies emerged. Murine mesotheliomas were dominated by myeloid cells, particularly macrophages, while anti-tumour lymphocytes were scarce, indicating failure to recapitulate the complex microenvironment of human MPM, which discouraged ICB experiments. However, multiplex staining of biopsies obtained before nivolumab administration revealed several differentiators of clinical response, including high CTL density within programmed death ligand 1 (PD-L1)+ tumours and tertiary lymphoid structures (TLS), especially mature TLS with distinctively zoned secondary follicles. TLSs also epitomised a 'hot' microenvironment, existing primarily in tumours with substantial anti-tumour infiltrates of B lymphocytes and naïve T lymphocytes. These latter findings shed light on mechanisms of ICB response in MPM in agreement with evidence from lung cancer and melanoma, further suggesting actionable candidate biomarkers for future patient stratification. Being the first histopathological data from ICB-treated MPM, and the first data implicating TLS in MPM drug response, these results should be validated in future patient cohorts and could be refined further when supplemented with connected 'omics data.
- Published
- 2023
- Full Text
- View/download PDF
5. Comparing anti-tumor and anti-self immunity in a patient with melanoma receiving immune checkpoint blockade
- Author
-
Shuming Chen, Tracee L. McMiller, Abha Soni, Farah Succaria, John-William Sidhom, Laura C. Cappelli, Livia A. Casciola-Rosen, Isaac R. Morales, Preethi Sankaran, Alan E. Berger, Julie Stein Deutsch, Qingfeng C. Zhu, Robert A. Anders, Jody E. Hooper, Drew M. Pardoll, Evan J. Lipson, Janis M. Taube, and Suzanne L. Topalian
- Subjects
COX-2 ,Gene expression profiling (GEP) ,Immune checkpoint blockade (ICB) ,Immune related adverse events (irAEs) ,Melanoma ,Rapid autopsy ,Medicine - Abstract
Abstract Background Tumor regression following immune checkpoint blockade (ICB) is often associated with immune-related adverse events (irAEs), marked by inflammation in non-cancerous tissues. This study was undertaken to investigate the functional relationship between anti-tumor and anti-self immunity, to facilitate irAE management while promoting anti-tumor immunity. Methods Multiple biopsies from tumor and inflamed tissues were collected from a patient with melanoma experiencing both tumor regression and irAEs on ICB, who underwent rapid autopsy. Immune cells infiltrating melanoma lesions and inflamed normal tissues were subjected to gene expression profiling with multiplex qRT-PCR for 122 candidate genes. Subsequently, immunohistochemistry was conducted to assess the expression of 14 candidate markers of immune cell subsets and checkpoints. TCR-beta sequencing was used to explore T cell clonal repertoires across specimens. Results While genes involved in MHC I/II antigen presentation, IFN signaling, innate immunity and immunosuppression were abundantly expressed across specimens, irAE tissues over-expressed certain genes associated with immunosuppression (CSF1R, IL10RA, IL27/EBI3, FOXP3, KLRG1, SOCS1, TGFB1), including those in the COX-2/PGE2 pathway (IL1B, PTGER1/EP1 and PTGER4/EP4). Immunohistochemistry revealed similar proportions of immunosuppressive cell subsets and checkpoint molecules across samples. TCRseq did not indicate common TCR repertoires across tumor and inflammation sites, arguing against shared antigen recognition between anti-tumor and anti-self immunity in this patient. Conclusions This comprehensive study of a single patient with melanoma experiencing both tumor regression and irAEs on ICB explores the immune landscape across these tissues, revealing similarities between anti-tumor and anti-self immunity. Further, it highlights expression of the COX-2/PGE2 pathway, which is known to be immunosuppressive and potentially mediates ICB resistance. Ongoing clinical trials of COX-2/PGE2 pathway inhibitors targeting the major COX-2 inducer IL-1B, COX-2 itself, or the PGE2 receptors EP2 and EP4 present new opportunities to promote anti-tumor activity, but may also have the potential to enhance the severity of ICB-induced irAEs.
- Published
- 2024
- Full Text
- View/download PDF
6. Targeting of focal adhesion kinase enhances the immunogenic cell death of PEGylated liposome doxorubicin to optimize therapeutic responses of immune checkpoint blockade
- Author
-
Zhang, Baoyuan, Li, Ning, Gao, Jiaming, Zhao, Yuxi, Jiang, Jun, Xie, Shuang, Zhang, Cuiping, Zhang, Qingyu, Liu, Leo, Wang, Zaiqi, Ji, Dongmei, Wu, Lingying, and Ren, Ruibao
- Published
- 2024
- Full Text
- View/download PDF
7. MIF and CD74 as Emerging Biomarkers for Immune Checkpoint Blockade Therapy.
- Author
-
Fey, Rosalyn M., Nichols, Rebecca A., Tran, Thuy T., Vandenbark, Arthur A., and Kulkarni, Rajan P.
- Subjects
- *
TUMOR treatment , *DRUG toxicity , *LYMPHOKINES , *MELANOMA , *TUMOR markers , *IMMUNE checkpoint inhibitors , *AUTOIMMUNE diseases , *CYTOKINES , *CELL receptors , *DISEASE progression - Abstract
Simple Summary: While immune checkpoint blockade (ICB) therapy is used successfully to treat various cancers, many patients develop immune-related adverse side effects (irAEs), which can be severe enough to cause decreases in quality of life and, in some cases, may result in treatment cessation. Here, we review current studies investigating the potential utility of the molecules MIF and CD74 as predictive biomarkers for ICB response and irAE development. We also discuss evidence for the circadian expression of MIF. Finally, we aim to highlight areas where future research will be beneficial in establishing the value of MIF and CD74 as biomarkers of ICB response. Immune checkpoint blockade (ICB) therapy is used to treat a wide range of cancers; however, some patients are at risk of developing treatment resistance and/or immune-related adverse events (irAEs). Thus, there is a great need for the identification of reliable predictive biomarkers for response and toxicity. The cytokine MIF (macrophage migration inhibitory factor) and its cognate receptor CD74 are intimately connected with cancer progression and have previously been proposed as prognostic biomarkers for patient outcome in various cancers, including solid tumors such as malignant melanoma. Here, we assess their potential as predictive biomarkers for response to ICB therapy and irAE development. We provide a brief overview of their function and roles in the context of cancer and autoimmune disease. We also review the evidence showing that MIF and CD74 may be of use as predictive biomarkers of patient response to ICB therapy and irAE development. We also highlight that careful consideration is required when assessing the potential of serum MIF levels as a biomarker due to its reported circadian expression in human plasma. Finally, we suggest future directions for the establishment of MIF and CD74 as predictive biomarkers for ICB therapy and irAE development to guide further research in this field. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Comparing anti-tumor and anti-self immunity in a patient with melanoma receiving immune checkpoint blockade.
- Author
-
Chen, Shuming, McMiller, Tracee L., Soni, Abha, Succaria, Farah, Sidhom, John-William, Cappelli, Laura C., Casciola-Rosen, Livia A., Morales, Isaac R., Sankaran, Preethi, Berger, Alan E., Deutsch, Julie Stein, Zhu, Qingfeng C., Anders, Robert A., Hooper, Jody E., Pardoll, Drew M., Lipson, Evan J., Taube, Janis M., and Topalian, Suzanne L.
- Subjects
IMMUNE checkpoint proteins ,DRUG side effects ,IPILIMUMAB ,IMMUNITY ,T cell receptors ,GENE expression profiling ,BIOMARKERS - Abstract
Background: Tumor regression following immune checkpoint blockade (ICB) is often associated with immune-related adverse events (irAEs), marked by inflammation in non-cancerous tissues. This study was undertaken to investigate the functional relationship between anti-tumor and anti-self immunity, to facilitate irAE management while promoting anti-tumor immunity. Methods: Multiple biopsies from tumor and inflamed tissues were collected from a patient with melanoma experiencing both tumor regression and irAEs on ICB, who underwent rapid autopsy. Immune cells infiltrating melanoma lesions and inflamed normal tissues were subjected to gene expression profiling with multiplex qRT-PCR for 122 candidate genes. Subsequently, immunohistochemistry was conducted to assess the expression of 14 candidate markers of immune cell subsets and checkpoints. TCR-beta sequencing was used to explore T cell clonal repertoires across specimens. Results: While genes involved in MHC I/II antigen presentation, IFN signaling, innate immunity and immunosuppression were abundantly expressed across specimens, irAE tissues over-expressed certain genes associated with immunosuppression (CSF1R, IL10RA, IL27/EBI3, FOXP3, KLRG1, SOCS1, TGFB1), including those in the COX-2/PGE2 pathway (IL1B, PTGER1/EP1 and PTGER4/EP4). Immunohistochemistry revealed similar proportions of immunosuppressive cell subsets and checkpoint molecules across samples. TCRseq did not indicate common TCR repertoires across tumor and inflammation sites, arguing against shared antigen recognition between anti-tumor and anti-self immunity in this patient. Conclusions: This comprehensive study of a single patient with melanoma experiencing both tumor regression and irAEs on ICB explores the immune landscape across these tissues, revealing similarities between anti-tumor and anti-self immunity. Further, it highlights expression of the COX-2/PGE2 pathway, which is known to be immunosuppressive and potentially mediates ICB resistance. Ongoing clinical trials of COX-2/PGE2 pathway inhibitors targeting the major COX-2 inducer IL-1B, COX-2 itself, or the PGE2 receptors EP2 and EP4 present new opportunities to promote anti-tumor activity, but may also have the potential to enhance the severity of ICB-induced irAEs. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Review immune response of targeting CD39 in cancer
- Author
-
Yao Liu, Zhongliang Li, Xiaoguang Zhao, Jing Xiao, Jiacheng Bi, Xian-Yang Li, Guokai Chen, and Ligong Lu
- Subjects
CD39 ,Adenosine pathway ,Immune checkpoint blockade (ICB) ,PD-1 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract The ATP-adenosine pathway has emerged as a promising target for cancer therapy, but challenges remain in achieving effective tumor control. Early research focused on blocking the adenosine generating enzyme CD73 and the adenosine receptors A2AR or A2BR in cancer. However, recent studies have shown that targeting CD39, the rate-limiting ecto-enzyme of the ATP-adenosine pathway, can provide more profound anti-tumor efficacy by reducing immune-suppressive adenosine accumulation and increasing pro-inflammatory ATP levels. In addition, combining CD39 blocking antibody with PD-1 immune checkpoint therapy may have synergistic anti-tumor effects and improve patient survival. This review will discuss the immune components that respond to CD39 targeting in the tumor microenvironment. Targeting CD39 in cancer has been shown to not only decrease adenosine levels in the tumor microenvironment (TME), but also increase ATP levels. Additionally, targeting CD39 can limit the function of Treg cells, which are known to express high levels of CD39. With phase I clinical trials of CD39 targeting currently underway, further understanding and rational design of this approach for cancer therapy are expected.
- Published
- 2023
- Full Text
- View/download PDF
10. Metabolic interventions combined with CTLA-4 and PD-1/PD-L1 blockade for the treatment of tumors: mechanisms and strategies.
- Author
-
Liao, Liming, Xu, Huilin, Zhao, Yuhan, and Zheng, Xiaofeng
- Abstract
Immunotherapies based on immune checkpoint blockade (ICB) have significantly improved patient outcomes and offered new approaches to cancer therapy over the past decade. To date, immune checkpoint inhibitors (ICIs) of CTLA-4 and PD-1/PD-L1 represent the main class of immunotherapy. Blockade of CTLA-4 and PD-1/PD-L1 has shown remarkable efficacy in several specific types of cancers, however, a large subset of refractory patients presents poor responsiveness to ICB therapy; and the underlying mechanism remains elusive. Recently, numerous studies have revealed that metabolic reprogramming of tumor cells restrains immune responses by remodeling the tumor microenvironment (TME) with various products of metabolism, and combination therapies involving metabolic inhibitors and ICIs provide new approaches to cancer therapy. Nevertheless, a systematic summary is lacking regarding the manner by which different targetable metabolic pathways regulate immune checkpoints to overcome ICI resistance. Here, we demonstrate the generalized mechanism of targeting cancer metabolism at three crucial immune checkpoints (CTLA-4, PD-1, and PD-L1) to influence ICB therapy and propose potential combined immunotherapeutic strategies co-targeting tumor metabolic pathways and immune checkpoints. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Immune checkpoint inhibition mediated with liposomal nanomedicine for cancer therapy
- Author
-
Guang-Long Ma and Wei-Feng Lin
- Subjects
Liposome ,Exosome ,Immune checkpoint blockade (ICB) ,Medicine (General) ,R5-920 ,Military Science - Abstract
Abstract Immune checkpoint blockade (ICB) therapy for cancer has achieved great success both in clinical results and on the market. At the same time, success drives more attention from scientists to improve it. However, only a small portion of patients are responsive to this therapy, and it comes with a unique spectrum of side effects termed immune-related adverse events (irAEs). The use of nanotechnology could improve ICBs’ delivery to the tumor, assist them in penetrating deeper into tumor tissues and alleviate their irAEs. Liposomal nanomedicine has been investigated and used for decades, and is well-recognized as the most successful nano-drug delivery system. The successful combination of ICB with liposomal nanomedicine could help improve the efficacy of ICB therapy. In this review, we highlighted recent studies using liposomal nanomedicine (including new emerging exosomes and their inspired nano-vesicles) in associating ICB therapy.
- Published
- 2023
- Full Text
- View/download PDF
12. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment
- Author
-
Zhifei Gao, Yifeng Bai, Anqi Lin, Aimin Jiang, Chaozheng Zhou, Quan Cheng, Zaoqu Liu, Xin Chen, Jian Zhang, and Peng Luo
- Subjects
γδT cells ,Immune checkpoint molecules ,Immune checkpoint inhibitors (ICIs) ,Immune checkpoint blockade (ICB) ,Checkpoint inhibitor (CPI) ,Tumor microenvironment (TME) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract As a nontraditional T-cell subgroup, γδT cells have gained popularity in the field of immunotherapy in recent years. They have extraordinary antitumor potential and prospects for clinical application. Immune checkpoint inhibitors (ICIs), which are efficacious in tumor patients, have become pioneer drugs in the field of tumor immunotherapy since they were incorporated into clinical practice. In addition, γδT cells that have infiltrated into tumor tissues are found to be in a state of exhaustion or anergy, and there is upregulation of many immune checkpoints (ICs) on their surface, suggesting that γδT cells have a similar ability to respond to ICIs as traditional effector T cells. Studies have shown that targeting ICs can reverse the dysfunctional state of γδT cells in the tumor microenvironment (TME) and exert antitumor effects by improving γδT-cell proliferation and activation and enhancing cytotoxicity. Clarification of the functional state of γδT cells in the TME and the mechanisms underlying their interaction with ICs will solidify ICIs combined with γδT cells as a good treatment option.
- Published
- 2023
- Full Text
- View/download PDF
13. Drug-loaded microbubble delivery system to enhance PD-L1 blockade immunotherapy with remodeling immune microenvironment
- Author
-
Jun Zheng, Ju Huang, Liang Zhang, Mengna Wang, Lihong Xu, Xiaoyun Dou, Xiaojing Leng, Mingxiao Fang, Yang Sun, and Zhigang Wang
- Subjects
Ultrasound-targeted microbubble destruction (UTMD) ,Immune checkpoint blockade (ICB) ,Docetaxel (DTX) ,PD-L1 ,Tumor immunosuppressive microenvironment ,Medical technology ,R855-855.5 - Abstract
Abstract Background Although programmed cell death protein 1 (PD-1)/ programmed cell death-ligand protein 1 (PD-L1) checkpoint blockade immunotherapy demonstrates great promise in cancer treatment, poor infiltration of T cells resulted from tumor immunosuppressive microenvironment (TIME) and insufficient accumulation of anti-PD-L1 (αPD-L1) in tumor sites diminish the immune response. Herein, we reported a drug-loaded microbubble delivery system to overcome these obstacles and enhance PD-L1 blockade immunotherapy. Methods Docetaxel (DTX) and imiquimod (R837)-loaded microbubbles (RD@MBs) were synthesized via a typical rotary evaporation method combined with mechanical oscillation. The targeted release of drugs was achieved by using the directional "bursting" capability of ultrasound-targeted microbubble destruction (UTMD) technology. The antitumor immune response by RD@MBs combining αPD-L1 were evaluated on 4T1 and CT26 tumor models. Results The dying tumor cells induced by DTX release tumor-associated antigens (TAAs), together with R837, promoted the activation, proliferation and recruitment of T cells. Besides, UTMD technology and DTX enhanced the accumulation of αPD-L1 in tumor sites. Moreover, RD@MBs remolded TIME, including the polarization of M2-phenotype tumor-associated macrophages (TAMs) to M1-phenotype, and reduction of myeloid-derived suppressor cells (MDSCs). The RD@MBs + αPD-L1 synergistic therapy not only effectively inhibited the growth of primary tumors, but also significantly inhibited the mimic distant tumors as well as lung metastases. Conclusion PD-L1 blockade immunotherapy was enhanced by RD@MBs delivery system.
- Published
- 2023
- Full Text
- View/download PDF
14. PCSK9 facilitates melanoma pathogenesis via a network regulating tumor immunity
- Author
-
Yan Gu, Xiaozeng Lin, Ying Dong, Geoffrey Wood, Nabil G. Seidah, Geoff Werstuck, Pierre Major, Michael Bonert, Anil Kapoor, and Damu Tang
- Subjects
PCSK9 ,Melanoma ,Tumorigenesis ,Immune checkpoint blockade (ICB) ,Biomarkers of ICB ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background PCSK9 regulates cholesterol homeostasis and promotes tumorigenesis. However, the relevance of these two actions and the mechanisms underlying PCSK9’s oncogenic roles in melanoma and other cancers remain unclear. Methods PCSK9’s association with melanoma was analysed using the TCGA dataset. Empty vector (EV), PCSK9, gain-of-function (D374Y), and loss-of-function (Q152H) PCSK9 mutant were stably-expressed in murine melanoma B16 cells and studied for impact on B16 cell-derived oncogenesis in vitro and in vivo using syngeneic C57BL/6 and Pcsk9 −/− mice. Intratumoral accumulation of cholesterol was determined. RNA-seq was performed on individual tumor types. Differentially-expressed genes (DEGs) were derived from the comparisons of B16 PCSK9, B16 D374Y, or B16 Q152H tumors to B16 EV allografts and analysed for pathway alterations. Results PCSK9 expression and its network negatively correlated with the survival probability of patients with melanoma. PCSK9 promoted B16 cell proliferation, migration, and growth in soft agar in vitro, formation of tumors in C57BL/6 mice in vivo, and accumulation of intratumoral cholesterol in a manner reflecting its regulation of the low-density lipoprotein receptor (LDLR): Q152H, EV, PCSK9, and D374Y. Tumor-associated T cells, CD8 + T cells, and NK cells were significantly increased in D374Y tumors along with upregulations of multiple immune checkpoints, IFNγ, and 143 genes associated with T cell dysfunction. Overlap of 36 genes between the D374Y DEGs and the PCSK9 DEGs predicted poor prognosis of melanoma and resistance to immune checkpoint blockade (ICB) therapy. CYTH4, DENND1C, AOAH, TBC1D10C, EPSTI1, GIMAP7, and FASL (FAS ligand) were novel predictors of ICB therapy and displayed high level of correlations with multiple immune checkpoints in melanoma and across 30 human cancers. We observed FAS ligand being among the most robust biomarkers of ICB treatment and constructed two novel and effective multigene panels predicting response to ICB therapy. The profiles of allografts produced by B16 EV, PCSK9, D374Y, and Q152H remained comparable in C57BL/6 and Pcsk9 −/− mice. Conclusions Tumor-derived PCSK9 plays a critical role in melanoma pathogenesis. PCSK9’s oncogenic actions are associated with intratumoral cholesterol accumulation. PCSK9 systemically affects the immune system, contributing to melanoma immune evasion. Novel biomarkers derived from the PCSK9-network effectively predicted ICB therapy responses.
- Published
- 2023
- Full Text
- View/download PDF
15. A Comprehensive Benchmark of Transcriptomic Biomarkers for Immune Checkpoint Blockades.
- Author
-
Kang, Hongen, Zhu, Xiuli, Cui, Ying, Xiong, Zhuang, Zong, Wenting, Bao, Yiming, and Jia, Peilin
- Subjects
- *
BIOMARKERS , *IMMUNE checkpoint inhibitors , *HEALTH status indicators , *TREATMENT effectiveness , *GENE expression profiling , *RESEARCH funding , *SURVIVAL analysis (Biometry) , *TUMORS , *IMMUNOTHERAPY - Abstract
Simple Summary: Immune checkpoint blockades (ICBs) therapy has produced durable clinical responses in many cancer types, but only a fraction of patients can benefit from ICB treatment. Previous studies have reported multiple transcriptomic biomarkers to predict ICB responses and improve treatment precision in various cancer types. However, a timely and unbiased assessment of these biomarkers has yet to be conducted due to the lack of large-scale uniformly curated ICB-treated datasets. To address the needs, we developed ICB-Portal, a comprehensive resource about ICB including RNA-seq data of 29 datasets from public sources and standardized metadata of each study through a uniform pre-processing, 48 biomarker scores associated with ICB response, results of a systematic benchmark analysis evaluating the efficacy, and generalization ability for each biomarker in various scenarios such as different cancer types, anti-bodies, biopsy time, and combinatory treatments with other drugs by a standardized bioinformatics workflow and an online benchmark platform. Immune checkpoint blockades (ICBs) have revolutionized cancer therapy by inducing durable clinical responses, but only a small percentage of patients can benefit from ICB treatments. Many studies have established various biomarkers to predict ICB responses. However, different biomarkers were found with diverse performances in practice, and a timely and unbiased assessment has yet to be conducted due to the complexity of ICB-related studies and trials. In this study, we manually curated 29 published datasets with matched transcriptome and clinical data from more than 1400 patients, and uniformly preprocessed these datasets for further analyses. In addition, we collected 39 sets of transcriptomic biomarkers, and based on the nature of the corresponding computational methods, we categorized them into the gene-set-like group (with the self-contained design and the competitive design, respectively) and the deconvolution-like group. Next, we investigated the correlations and patterns of these biomarkers and utilized a standardized workflow to systematically evaluate their performance in predicting ICB responses and survival statuses across different datasets, cancer types, antibodies, biopsy times, and combinatory treatments. In our benchmark, most biomarkers showed poor performance in terms of stability and robustness across different datasets. Two scores (TIDE and CYT) had a competitive performance for ICB response prediction, and two others (PASS-ON and EIGS_ssGSEA) showed the best association with clinical outcome. Finally, we developed ICB-Portal to host the datasets, biomarkers, and benchmark results and to implement the computational methods for researchers to test their custom biomarkers. Our work provided valuable resources and a one-stop solution to facilitate ICB-related research. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Tertiary Lymphoid Structures Are Associated with a Favorable Prognosis in High-Grade Serous Ovarian Cancer Patients.
- Author
-
Zhang, Ke, Xie, Xiao, Zou, Li-Hao, and Guo, Sui-Qun
- Abstract
There was accumulating evidence indicating that tertiary lymphoid structures (TLSs) were strongly associated with improved survival and clinical outcome in several solid tumors. In this study, we intended to assess the presence of TLSs and their potential clinical significance in high-grade serous ovarian cancer (HGSOC). TCGA (The Cancer Genome Atlas) cohort included RNA-seq data of 376 HGSOC patients, of which 74 patients included available hematoxylin-eosin (H&E) sections; GEO (Gene Expression Omnibus) cohort, GSE140082, included microarray data of 212 HGSOC patients. TLSs were counted by pathological sections, and the relative abundance of TLSs was assessed by the unsupervised consensus clustering of 12-chemokine transcriptome signatures. The potential associations between TLSs and clinical prognosis, tumor microenvironment (TME), and immunotherapy response of HGSOC were further performed based on transcriptome data. In the H&E sections of HGSOC, TLSs were predominantly located in the stroma and invasive margin of the tumor. Pathological counting results suggested that the expression of 12 chemokines was significantly higher in samples abundant with TLSs than that in the lack of TLSs. Consensus clustering of both TCGA and GEO cohorts divided HGSOC patients into two clusters with different TLSs abundance: low- and high-TLSs. Based on transcriptome analysis, the high-TLS cluster was characterized by better clinical prognosis, a higher degree of immune infiltration, more biological pathways, higher tumor mutational burden score, and higher expression of immune checkpoints. In conclusion, TLSs strongly correlated with the immune-responsive microenvironment and remained a favorable prognostic factor independent of other clinical characteristics in HGSOC. The presence of TLSs was also associated with a potentially favorable response to immune checkpoint blockade (ICB) therapy in HGSOC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Review immune response of targeting CD39 in cancer.
- Author
-
Liu, Yao, Li, Zhongliang, Zhao, Xiaoguang, Xiao, Jing, Bi, Jiacheng, Li, Xian-Yang, Chen, Guokai, and Lu, Ligong
- Subjects
ADENOSINES ,IMMUNE response ,REGULATORY T cells ,IMMUNE checkpoint proteins ,TUMOR microenvironment ,OVERALL survival ,PROGRAMMED cell death 1 receptors - Abstract
The ATP-adenosine pathway has emerged as a promising target for cancer therapy, but challenges remain in achieving effective tumor control. Early research focused on blocking the adenosine generating enzyme CD73 and the adenosine receptors A2AR or A2BR in cancer. However, recent studies have shown that targeting CD39, the rate-limiting ecto-enzyme of the ATP-adenosine pathway, can provide more profound anti-tumor efficacy by reducing immune-suppressive adenosine accumulation and increasing pro-inflammatory ATP levels. In addition, combining CD39 blocking antibody with PD-1 immune checkpoint therapy may have synergistic anti-tumor effects and improve patient survival. This review will discuss the immune components that respond to CD39 targeting in the tumor microenvironment. Targeting CD39 in cancer has been shown to not only decrease adenosine levels in the tumor microenvironment (TME), but also increase ATP levels. Additionally, targeting CD39 can limit the function of Treg cells, which are known to express high levels of CD39. With phase I clinical trials of CD39 targeting currently underway, further understanding and rational design of this approach for cancer therapy are expected. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
18. Loss of BLK expression as a potential predictor of poor prognosis and immune checkpoint blockade response in NSCLC and contribute to tumor progression
- Author
-
Yingqi Xu, Jianlin Xu, Rong Qiao, Hua Zhong, Jinjing Xia, and Runbo Zhong
- Subjects
Non-small cell lung cancer (NSCLC) ,Immune checkpoint blockade (ICB) ,BLK ,prognosis ,Tumor growth ,RNA sequencing ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Background: Immune checkpoint blockade (ICB) has been proved to have significant anti-tumor effect in the clinical treatment of non-small cell lung cancer (NSCLC). Therefore, biomarkers predicting ICB response can provide better treatment for patients with NSCLC. Methods: Differential expression genes (DEGs) were identified by ImmuCellAI database. Copy number alteration (CNA) was analyzed by cBioPortal. The predicted efficiency of 4 genes on cancer immunotherapy was assessed by ROC analysis. The survival value of BLK was analyzed by Kaplan-Meier plotter and Prognoscan analysis. Clinical significance of BLK IHC-TMA score in NSCLC was also explored. The CCK-8 assay, wound healing assay, western blot assay in vitro and subcutaneous xenograft experiments in vivo were used for investigating the functions of BLK. The RNA-sequencing were performed to screen BLK regulated genes and conducted for GO/KEGG enrichment analysis. The transcriptional regulatory factor of BLK promoter region was predicted by ChIP-seq analysis. Results: 39 common DEGs between ICB Response (R) group and No Response (NR) group with NSCLC were identified, in which the CNA frequency of BLK deletion (> 6%) was found. The predicted efficiency of BLK on immunotherapy was performed best in NSCLC (AUC>0.7). Low expression of BLK was related to NSCLC with significantly poor prognosis. BLK overexpression can inhibit growth of NSCLC via activating apoptosis pathway, inhibiting the G2M checkpoint and Glycolysis pathway. The enrichment analysis indicated that BLK regulated genes related to oncogenic potential in NSCLC. Besides, BLK expression was inhibited via H3K27me3 modification in A549 and H1299 cells. BLK mRNA level was negatively correlated with methylation and positively correlated with the tumor purity in NSCLC. Conclusion: Our study provides strong evidence that low expression of BLK may serve as a biomarker for poor prognosis in NSCLC, while response to ICB therapy and contributes to NSCLC tumor progression.
- Published
- 2023
- Full Text
- View/download PDF
19. Immune checkpoint inhibition mediated with liposomal nanomedicine for cancer therapy.
- Author
-
Ma, Guang-Long and Lin, Wei-Feng
- Subjects
IMMUNE checkpoint inhibitors ,NANOMEDICINE ,DRUG side effects ,CANCER treatment ,IMMUNE checkpoint proteins ,IPILIMUMAB - Abstract
Immune checkpoint blockade (ICB) therapy for cancer has achieved great success both in clinical results and on the market. At the same time, success drives more attention from scientists to improve it. However, only a small portion of patients are responsive to this therapy, and it comes with a unique spectrum of side effects termed immune-related adverse events (irAEs). The use of nanotechnology could improve ICBs' delivery to the tumor, assist them in penetrating deeper into tumor tissues and alleviate their irAEs. Liposomal nanomedicine has been investigated and used for decades, and is well-recognized as the most successful nano-drug delivery system. The successful combination of ICB with liposomal nanomedicine could help improve the efficacy of ICB therapy. In this review, we highlighted recent studies using liposomal nanomedicine (including new emerging exosomes and their inspired nano-vesicles) in associating ICB therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Gamma delta T-cell-based immune checkpoint therapy: attractive candidate for antitumor treatment.
- Author
-
Gao, Zhifei, Bai, Yifeng, Lin, Anqi, Jiang, Aimin, Zhou, Chaozheng, Cheng, Quan, Liu, Zaoqu, Chen, Xin, Zhang, Jian, and Luo, Peng
- Subjects
IMMUNE checkpoint proteins ,IMMUNE checkpoint inhibitors ,TUMOR microenvironment ,CLINICAL medicine ,T cells - Abstract
As a nontraditional T-cell subgroup, γδT cells have gained popularity in the field of immunotherapy in recent years. They have extraordinary antitumor potential and prospects for clinical application. Immune checkpoint inhibitors (ICIs), which are efficacious in tumor patients, have become pioneer drugs in the field of tumor immunotherapy since they were incorporated into clinical practice. In addition, γδT cells that have infiltrated into tumor tissues are found to be in a state of exhaustion or anergy, and there is upregulation of many immune checkpoints (ICs) on their surface, suggesting that γδT cells have a similar ability to respond to ICIs as traditional effector T cells. Studies have shown that targeting ICs can reverse the dysfunctional state of γδT cells in the tumor microenvironment (TME) and exert antitumor effects by improving γδT-cell proliferation and activation and enhancing cytotoxicity. Clarification of the functional state of γδT cells in the TME and the mechanisms underlying their interaction with ICs will solidify ICIs combined with γδT cells as a good treatment option. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Immunosuppression, immune escape, and immunotherapy in pancreatic cancer: focused on the tumor microenvironment.
- Author
-
Zhu, Yu-Heng, Zheng, Jia-Hao, Jia, Qin-Yuan, Duan, Zong-Hao, Yao, Hong-Fei, Yang, Jian, Sun, Yong-Wei, Jiang, Shu-Heng, Liu, De-Jun, and Huo, Yan-Miao
- Subjects
- *
PANCREATIC cancer , *TUMOR microenvironment , *IMMUNOTHERAPY , *PANCREATIC duct , *IMMUNOSUPPRESSION - Abstract
Pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is characterized by poor treatment response and low survival time. The current clinical treatment for advanced PDAC is still not effective. In recent years, the research and application of immunotherapy have developed rapidly and achieved substantial results in many malignant tumors. However, the translational application in PDAC is still far from satisfactory and needs to be developed urgently. To carry out the study of immunotherapy, it is necessary to fully decipher the immune characteristics of PDAC. This review summarizes the recent progress of the tumor microenvironment (TME) of PDAC and highlights its link with immunotherapy. We describe the molecular cues and corresponding intervention methods, collate several promising targets and progress worthy of further study, and put forward the importance of integrated immunotherapy to provide ideas for future research of TME and immunotherapy of PDAC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. CXCR4 表达联合 BRAF 突变分析对筛选皮肤 黑色素瘤免疫治疗人群的作用.
- Author
-
戴赛林, 邵甲云, 雪 燕, and 郑 洁
- Abstract
Objective To find suitable markers combined with BRAF mutation analysis to screen out skin cutaneous melanoma (SKCM) patients that are suitable for immunotherapy. Methods We acquired the database of SKCM from the The Cancer Genome Atlas (TCGA) and divided the patients (n=470) into wild-type group (n=230) and mutation group (n=235). Limma package of R software was used to study the expression of mRNAs in the two groups. Differentially expressed genes (DEGs) were screened out by fold-change values and adjusted P. We performed enrichment analysis of DEGs via the Gene Ontology (GO) database. String database showed that CXCR4 might interact closely with TP53 and IL18. Pearson correlation analyses were used to describe the correlation between CXCR4 and immune checkpoint. Then we used Spearman's correlation analysis to describe the correlation between CXCR4 expression and microsatellite instability (MSI). Wilcoxon test was applied to analyze the difference in CXCR4 expression between the BRAF mutation group and the non-mutation group. Lastly, the expression levels of SIGLEC15, TIGIT, CD274, HAVCR2, PDCD1, CTLA4, LAG3, and PDCD1LG2 were implemented by packages ggplot2 and pheatmap in R foundation. Moreover, the potential immune checkpoint blockade (ICB) response of each subgroup was predicted with tumor immune dysfunction and exclusion (TIDE) algorithm. Results BRAF gene was widely mutated in SKCM, and DEGs caused by BRAF mutation were mainly concentrated in the IL18 and TP53 pathways. Moreover, CXCR4 was a chemokine closely associated with tumors, and had an expression regulatory network with IL18 and TP53. We found that the expression of CXCR4 remarkably increased in tumor tissues of SKCM, especially in mutation group. We further analyzed the correlation between CXCR4 and immune checkpoints. Interestingly, CXCR4 positively correlated with HAVCR2, PDCD1, PDCD1LG2 and TIGIT. Furtherly, combining BRAF mutation analysis with CXCR4 expression, we divided SKCM patients into 4 subgroups, which were BRAFWT/CXCR4-high (G1), BRAFWT/CXCR4-low (G2), BRAFMUT/CXCR4-high (G3) and BRAFMUT/CXCR4-low (G4). We selected the subgroup which suited for immunotherapy via analyzing the MSI score and expressions of the immune checkpoint, and then predicting immune response. Notably, the subgroup with lower CXCR4 expression was accompanied by higher MSI instability, lower immune checkpoint expression and lower TIDE score. Namely, G2 is the subgroup most suitable for ICB treatment. Conclusion CXCR4 is hopeful to be a potential biomarker for monitoring curative effects. Combining with BRAF detection, it provides a theoretical basis for screening out appropriate SKCM patients to receive ICB treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. PCSK9 facilitates melanoma pathogenesis via a network regulating tumor immunity.
- Author
-
Gu, Yan, Lin, Xiaozeng, Dong, Ying, Wood, Geoffrey, Seidah, Nabil G., Werstuck, Geoff, Major, Pierre, Bonert, Michael, Kapoor, Anil, and Tang, Damu
- Subjects
MELANOMA ,IMMUNE checkpoint proteins ,LIPOPROTEIN receptors ,KILLER cells ,LABORATORY mice ,T cells - Abstract
Background: PCSK9 regulates cholesterol homeostasis and promotes tumorigenesis. However, the relevance of these two actions and the mechanisms underlying PCSK9's oncogenic roles in melanoma and other cancers remain unclear. Methods: PCSK9's association with melanoma was analysed using the TCGA dataset. Empty vector (EV), PCSK9, gain-of-function (D374Y), and loss-of-function (Q152H) PCSK9 mutant were stably-expressed in murine melanoma B16 cells and studied for impact on B16 cell-derived oncogenesis in vitro and in vivo using syngeneic C57BL/6 and Pcsk9
−/− mice. Intratumoral accumulation of cholesterol was determined. RNA-seq was performed on individual tumor types. Differentially-expressed genes (DEGs) were derived from the comparisons of B16 PCSK9, B16 D374Y, or B16 Q152H tumors to B16 EV allografts and analysed for pathway alterations. Results: PCSK9 expression and its network negatively correlated with the survival probability of patients with melanoma. PCSK9 promoted B16 cell proliferation, migration, and growth in soft agar in vitro, formation of tumors in C57BL/6 mice in vivo, and accumulation of intratumoral cholesterol in a manner reflecting its regulation of the low-density lipoprotein receptor (LDLR): Q152H, EV, PCSK9, and D374Y. Tumor-associated T cells, CD8 + T cells, and NK cells were significantly increased in D374Y tumors along with upregulations of multiple immune checkpoints, IFNγ, and 143 genes associated with T cell dysfunction. Overlap of 36 genes between the D374Y DEGs and the PCSK9 DEGs predicted poor prognosis of melanoma and resistance to immune checkpoint blockade (ICB) therapy. CYTH4, DENND1C, AOAH, TBC1D10C, EPSTI1, GIMAP7, and FASL (FAS ligand) were novel predictors of ICB therapy and displayed high level of correlations with multiple immune checkpoints in melanoma and across 30 human cancers. We observed FAS ligand being among the most robust biomarkers of ICB treatment and constructed two novel and effective multigene panels predicting response to ICB therapy. The profiles of allografts produced by B16 EV, PCSK9, D374Y, and Q152H remained comparable in C57BL/6 and Pcsk9−/− mice. Conclusions: Tumor-derived PCSK9 plays a critical role in melanoma pathogenesis. PCSK9's oncogenic actions are associated with intratumoral cholesterol accumulation. PCSK9 systemically affects the immune system, contributing to melanoma immune evasion. Novel biomarkers derived from the PCSK9-network effectively predicted ICB therapy responses. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
24. Immune responses to SARS-CoV-2 in vaccinated patients receiving checkpoint blockade immunotherapy for cancer.
- Author
-
Piening, Alexander, Ebert, Emily, Khojandi, Niloufar, Alspach, Elise, and Teague, Ryan M.
- Subjects
IMMUNE response ,VACCINATION ,IMMUNOTHERAPY ,SARS-CoV-2 ,IMMUNE checkpoint proteins - Abstract
Vaccination against SARS-CoV-2 has been successful in protecting patients with cancer from severe infections, but how immune responses against COVID-19 vaccination interact with those elicited during cancer immunotherapy has not been fully described. Immune checkpoint blockade (ICB) disrupts inhibitory pathways in immune cells to improve function and induce tumor immunity but can often cause serious immune related adverse events (IRAEs). Because COVID-19 vaccination and ICB both boost immune responses, it is imperative to understand if combining these regimens causes synergistic enhancement of the immune system. Specifically, whether ICB impacts anti-vaccine immunity in previously vaccinated patients is important since a large percentage of newly diagnosed cancer patients eligible for immunotherapy will have already been vaccinated against COVID-19. To address this, we investigated the influence of ICB on SARS-CoV-2-spike protein (SP) antibody titers and T cell responses in cancer patients previously vaccinated against COVID-19. Human blood samples were collected from 29 vaccinated patients and 12 unvaccinated control patients at baseline (prior to ICB) and following two rounds of ICB infusion. Anti-SARS-CoV-2-SP IgG titers and T cell responses were quantified. Compared to responses at baseline, there was no significant difference in these immune responses after immunotherapy in vaccinated individuals (P=0.4583, P=0.4571, respectively). We interpret these results as evidence that ICB immunotherapy does not significantly enhance SARS-CoV-2-specific antibody titers or T cell responses. Although our study lacks corresponding IRAE rates, the results provide humoral and cellular immunological data that support recent reports documenting the clinical safety and efficacy of COVID-19 vaccination in patients receiving ICB. Additional longitudinal prospective studies, such as the VOICE study (ClinicalTrials.gov identifier NCT04715438) and CAPTURE study (ClinicalTrials.gov identifier NCT03226886), are warranted and will provide broader safety and immunological data defining the effect of systemic cancer therapies on COVID-19 immunity. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. Overexpression of SMS in the tumor microenvironment is associated with immunosuppression in hepatocellular carcinoma.
- Author
-
Lin Xiang, Longhuan Piao, Dong Wang, and Li-Feng-Rong Qi
- Subjects
HEPATOCELLULAR carcinoma ,TUMOR microenvironment ,IMMUNE checkpoint proteins ,TH2 cells ,GENETIC overexpression - Abstract
Disorders of polyamine metabolism may contribute to the development of hepatocellular carcinoma (HCC), but the precise mechanism remains unknown. This study reports that spermine synthase (SMS), an enzyme involved in polyamine biosynthesis, is overexpressed in HCC and not associated with hepatitis virus infection in HCC patients. The results of analyzing the clinical data of HCC patients showed that SMS level as a categorical dependent variable was related to clinicopathological features of poor prognosis. Furthermore, the Kaplan-Meier survival analysis and ROC curve indicated that increased SMS level is associated with poor survival rate in HCC and may be a potential biomarker to discriminate HCC tissues. However, SMS overexpression limited the therapeutic effect of immune checkpoint blockade (ICB), which seemed to be related to the immunosuppressive effect of the HCC immune microenvironment formed by higher mRNA transcript levels of immune checkpoints and higher infiltration levels of immunosuppressive cells. In samples with high and low SMS expression, functional enrichment analysis of the differentially expressed genes (DEGs) showed that SMS may be linked to the occurrence and development of HCC by affecting a variety of immune-related pathways, such as Intestinal immune network for IgA production, Fc gamma R-mediated phagocytosis, Antigen processing and presentation, Th1 and Th2 cell differentiation. Subsequently, analysis of the co-expression network of SMS in the liver hepatocellular carcinoma (LIHC) cohort revealed that SMS has a broad impact on multiple important immune- and metabolic-related processes in HCC. In summary, SMS is a promising biomarker to differentiate the prognosis, immune characteristics, and holds promise as a potential target for ICB therapy to improve HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
26. Immune responses to SARS-CoV-2 in vaccinated patients receiving checkpoint blockade immunotherapy for cancer
- Author
-
Alexander Piening, Emily Ebert, Niloufar Khojandi, Elise Alspach, and Ryan M. Teague
- Subjects
immune checkpoint blockade (ICB) ,cancer ,COVID - 19 ,vaccination ,immunotherapy ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Vaccination against SARS-CoV-2 has been successful in protecting patients with cancer from severe infections, but how immune responses against COVID-19 vaccination interact with those elicited during cancer immunotherapy has not been fully described. Immune checkpoint blockade (ICB) disrupts inhibitory pathways in immune cells to improve function and induce tumor immunity but can often cause serious immune related adverse events (IRAEs). Because COVID-19 vaccination and ICB both boost immune responses, it is imperative to understand if combining these regimens causes synergistic enhancement of the immune system. Specifically, whether ICB impacts anti-vaccine immunity in previously vaccinated patients is important since a large percentage of newly diagnosed cancer patients eligible for immunotherapy will have already been vaccinated against COVID-19. To address this, we investigated the influence of ICB on SARS-CoV-2-spike protein (SP) antibody titers and T cell responses in cancer patients previously vaccinated against COVID-19. Human blood samples were collected from 29 vaccinated patients and 12 unvaccinated control patients at baseline (prior to ICB) and following two rounds of ICB infusion. Anti-SARS-CoV-2-SP IgG titers and T cell responses were quantified. Compared to responses at baseline, there was no significant difference in these immune responses after immunotherapy in vaccinated individuals (P=0.4583, P=0.4571, respectively). We interpret these results as evidence that ICB immunotherapy does not significantly enhance SARS-CoV-2-specific antibody titers or T cell responses. Although our study lacks corresponding IRAE rates, the results provide humoral and cellular immunological data that support recent reports documenting the clinical safety and efficacy of COVID-19 vaccination in patients receiving ICB. Additional longitudinal prospective studies, such as the VOICE study (ClinicalTrials.gov identifier NCT04715438) and CAPTURE study (ClinicalTrials.gov identifier NCT03226886), are warranted and will provide broader safety and immunological data defining the effect of systemic cancer therapies on COVID-19 immunity.
- Published
- 2022
- Full Text
- View/download PDF
27. Hsp90β inhibition upregulates interferon response and enhances immune checkpoint blockade therapy in murine tumors.
- Author
-
Rahmy, Sharif, Mishra, Sanket J., Murphy, Sean, Blagg, Brian S. J., and Xin Lu
- Subjects
IMMUNE checkpoint proteins ,HEAT shock proteins ,INTERFERONS ,PROSTATE cancer ,IMMUNE response ,TUMORS ,IPILIMUMAB - Abstract
Response resistance to the immune checkpoint blockade (ICB) immunotherapy remains a major clinical challenge that may be overcome through the rational combination of ICB and specific targeted therapeutics. One emerging combination strategy is based on sensitizing ICB-refractory tumors with antagonists of 90kD heat shock protein (Hsp90) that target all four isoforms. However, pan-Hsp90 inhibitors are limited by the modest efficacy, on-target and off-tumor toxicities, and induction of the heat shock response (HSR) that overrides the effect of Hsp90 inhibition. Recently, we developed Hsp90β-selective inhibitors that were cytotoxic to cancer cells but did not induce HSR in vitro. Here, we report that the Hsp90β inhibitor NDNB1182 downregulated CDK4 (an Hsp90β-dependent client protein) and induced the expression of endogenous retroviral elements and interferon-stimulated genes. In syngeneic mouse models of prostate cancer and breast cancer, NDNB1182 significantly augmented the efficacy of ICB therapy. Furthermore, NDNB1182 showed superior tolerability to the pan-Hsp90 inhibitor Ganetespib in mice. Our findings provide evidence that Hsp90β inhibition is a potentially effective and safe regimen to combine with ICB to treat immunotherapy-refractory solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. The Role of the Microbiome in Pancreatic Cancer.
- Author
-
Miyabayashi, Koji, Ijichi, Hideaki, and Fujishiro, Mitsuhiro
- Subjects
- *
PANCREATIC tumors , *IMMUNE checkpoint inhibitors , *CHRONIC diseases , *EARLY detection of cancer , *IMMUNE system , *DUCTAL carcinoma , *HUMAN microbiota , *PANCREATITIS , *PRECANCEROUS conditions , *DISEASE risk factors - Published
- 2022
- Full Text
- View/download PDF
29. Immunotherapy of targeting MDSCs in tumor microenvironment.
- Author
-
Hongshu Sui, Shengyi Dongye, Xiaocui Liu, Xinghua Xu, Li Wang, Jin, Christopher Q., Minhua Yao, Zhaoqing Gong, Daniel Jiang, Kexin Zhang, Yaling Liu, Hui Liu, Guomin Jiang, and Yanping Su
- Subjects
MYELOID-derived suppressor cells ,TUMOR microenvironment ,MYELOID cells ,T cells ,IMMUNE checkpoint proteins - Abstract
Myeloid-derived suppressor cells (MDSCs) are a group of heterogeneous cells which are abnormally accumulated during the differentiation of myeloid cells. Immunosuppression is the main functional feature of MDSCs, which inhibit T cell activity in the tumor microenvironment (TME) and promote tumoral immune escape. The main principle for immunotherapy is to modulate, restore, and remodel the plasticity and potential of immune system to have an effective anti-tumor response. In the TME, MDSCs are major obstacles to cancer immunotherapy through reducing the anti-tumor efficacy and making tumor cells more resistant to immunotherapy. Therefore, targeting MDSCs treatment becomes the priority of relevant studies and provides new immunotherapeutic strategy for cancer treatment. In this review, we mainly discuss the functions and mechanisms of MDSCs as well as their functional changes in the TME. Further, we review therapeutic effects of immunotherapy against MDSCs and potential breakthroughs regarding immunotherapy targeting MDSCs and immune checkpoint blockade (ICB) immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
30. Anti-PD1 Therapy Plus Whole-Brain Radiation Therapy May Prolong PFS in Selected Non–Small Cell Lung Cancer Patients with Brain Metastases: A Retrospective Study
- Author
-
Khan M, Zhao Z, Li X, and Liao G
- Subjects
brain metastasis (bm) ,whole brain radiation therapy (wbrt) ,non-small cell lung cancer (nsclc) ,immune checkpoint blockade (icb) ,combination (combined) therapy ,Medicine (General) ,R5-920 - Abstract
Muhammad Khan,1,2 Zhihong Zhao,3 Xianming Li,1 Guixiang Liao1 1Department of Oncology, Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, People’s Republic of China; 2Department of Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China; 3Department of Nephrology, Shenzhen People’s Hospital, Second Clinical Medicine Centre, Jinan University, Shenzhen, People’s Republic of ChinaCorrespondence: Guixiang Liao; Xianming LiDepartment of Radiation Oncology, Shenzhen People’s Hospital, First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, 518020, People’s Republic of ChinaEmail liaoguixiang@163.com; chenlhnfy@163.comBackground: Whole-brain radiotherapy (WBRT) remains an essential modality of treatment for brain metastases (BMs) derived from non-small cell lung cancer (NSCLC) patients and anti-PD-1 therapy has demonstrated intracranial responses in these patients. We aimed to evaluate if the combination of the two treatments could yield additive efficacy.Methods: A retrospective review of our institution’s database was carried out to identify NSCLC patients with BMs who had been treated with anti-PD1 therapy and/or WBRT between 2015 and 2020. Patient characteristics, main outcomes, including progression-free survival (PFS) and overall survival (OS), and factors affecting these outcomes were analyzed. SPSS 24 was used for statistical analysis. Appropriate statistical tests were employed according to the type of data.Results: Overall, 21 NSCLC BM patients were identified that had received WBRT. Of these, ten had been additionally treated with anti-PD1 therapy within 30 days of WBRT initiation. Median PFS was 3 (95% CI 0.8– 5.1) months with WBRT alone versus 11 (95% CI 6.3– 15.6) months with combined treatment. Risk of disease progression was 71% lower with the combined approach (HR 0.29, 95% CI 0.11– 0.80; p=0.016). A trend toward improved OS was also observed with the combined approach (HR 0.33, 95% CI 0.08– 1.12; p=0.107). Concurrent treatment (p=0.028) and male sex (p=0.052) were associated with improved PFS, while OS was associated only with age (p=0.02).Conclusion: Concurrent WBRT and anti-PD1 therapy may delay progression and improve survival in BM patients with confirmed EGFR- and ALK-negative NSCLC histology. Prospective studies are warranted to validate and elucidate on the additive effect of the two modalities.Keywords: brain metastasis, BM, whole-brain radiation therapy, WBRT, non–small cell lung cancer, NSCLC, immune checkpoint blockade, ICB, combination, combined therapy
- Published
- 2021
31. Editorial: Improvement of melanoma immune checkpoint blockade therapy with potential combinatorial regiments
- Author
-
Xueyan Wang, Xiaowei Liu, Willy Hugo, Lu Si, and Hubing Shi
- Subjects
immune checkpoint blockade (ICB) ,melanoma ,immune checkpoint inhibitors (ICIs) ,biomarker ,immune-related adverse events (irAEs) ,combination therapy ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2022
- Full Text
- View/download PDF
32. Hsp90β inhibition upregulates interferon response and enhances immune checkpoint blockade therapy in murine tumors
- Author
-
Sharif Rahmy, Sanket J. Mishra, Sean Murphy, Brian S. J. Blagg, and Xin Lu
- Subjects
heat shock protein 90 (hsp90) ,isoform-selective inhibitor ,immune checkpoint blockade (ICB) ,prostate cancer ,breast cancer ,CDK4/6 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Response resistance to the immune checkpoint blockade (ICB) immunotherapy remains a major clinical challenge that may be overcome through the rational combination of ICB and specific targeted therapeutics. One emerging combination strategy is based on sensitizing ICB-refractory tumors with antagonists of 90kD heat shock protein (Hsp90) that target all four isoforms. However, pan-Hsp90 inhibitors are limited by the modest efficacy, on-target and off-tumor toxicities, and induction of the heat shock response (HSR) that overrides the effect of Hsp90 inhibition. Recently, we developed Hsp90β-selective inhibitors that were cytotoxic to cancer cells but did not induce HSR in vitro. Here, we report that the Hsp90β inhibitor NDNB1182 downregulated CDK4 (an Hsp90β-dependent client protein) and induced the expression of endogenous retroviral elements and interferon-stimulated genes. In syngeneic mouse models of prostate cancer and breast cancer, NDNB1182 significantly augmented the efficacy of ICB therapy. Furthermore, NDNB1182 showed superior tolerability to the pan-Hsp90 inhibitor Ganetespib in mice. Our findings provide evidence that Hsp90β inhibition is a potentially effective and safe regimen to combine with ICB to treat immunotherapy-refractory solid tumors.
- Published
- 2022
- Full Text
- View/download PDF
33. Serine Protease Inhibitor Kazal Type 1, A Potential Biomarker for the Early Detection, Targeting, and Prediction of Response to Immune Checkpoint Blockade Therapies in Hepatocellular Carcinoma.
- Author
-
Jianlong Jia, Latai Ga, Yang Liu, Zhiyi Yang, Yue Wang, Xuanze Guo, Ruichen Ma, Ruonan Liu, Tianyou Li, Zeyao Tang, and Jun Wang
- Abstract
Background: We aimed to characterize serine protease inhibitor Kazal type 1 (SPINK1) as a gene signature for the early diagnosis, molecular targeting, and prediction of immune checkpoint blockade (ICB) treatment response of hepatocellular carcinoma (HCC).Methods: The transcriptomics, proteomics, and phenotypic analyses were performed separately or in combination.Results: We obtained the following findings on SPINK1. Firstly, in the transcriptomic training dataset, which included 279 stage I and II tumor samples (out of 1,884 stage I–IV HCC specimens) and 259 normal samples, significantly higher area under curve (AUC) values and increased integrated discrimination improvement (IDI) and net reclassification improvement (NRI) were demonstrated for HCC discrimination in SPINK1-associated models compared with those of alpha-fetoprotein (AFP). The calibration of both SPINK1-related curves fitted significantly better than that of AFP. In the two independent transcriptomic validation datasets, which included 201, 103 stage I-II tumor and 192, 169 paired non-tumor specimens, respectively, the obtained results were consistent with the above-described findings. In the proteomic training dataset, which included 98 stage I and II tumor and 165 normal tissue samples, the analyses also revealed better AUCs and increased IDI and NRI in the aforementioned SPINK1-associated settings. A moderate calibration was shown for both SPINK1-associated models relative to the poor results of AFP. Secondly, in the in vitro and/or in vivo murine models, the wet-lab experiments demonstrated that SPINK1 promoted the proliferation, clonal formation, migration, chemoresistance, anti-apoptosis, tumorigenesis, and metastasis of HCC cells, while the anti-SPINK1 antibody inhibited the growth of the cells, suggesting that SPINK1 has “tumor marker” and “targetable” characteristics in the management of HCC. The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses revealed that SPINK1 was engaged in immunity-related pathways, including T-cell activation. Thirdly, in the transcriptomic analyses of the 368 HCC specimens from The Cancer Genome Atlas (TCGA) cohort, the high abundance of SPINK1 was positively correlated with the high levels of activated tumor-infiltrating CD4
+ and CD8+ T lymphocytes and dendritic and natural killer cells, while there were also positive correlations between SPINK1 and immune checkpoints, including PD-1, LAG-3, TIM-3, TIGIT, HAVCR2, and CTLA-4. The ESTIMATE algorithm calculated positive correlations between SPINK1 and the immune and ESTIMATE scores, suggesting a close correlation between SPINK1 and the immunogenic microenvironment within HCC tissues, which may possibly help in predicting the response of patients to ICB therapy.Conclusions: SPINK1 could be a potential biomarker for the early detection, targeted therapy, and prediction of ICB treatment response in the management of HCC. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
34. Dose-Dependent Effect of Tumor Mutation Burden on Cancer Prognosis Following Immune Checkpoint Blockade: Causal Implications.
- Author
-
Zheng, Ming
- Subjects
IMMUNE checkpoint proteins ,CANCER prognosis ,HEAD & neck cancer - Abstract
High-TMB (TMB-H) and low-TMB (TMB-L) Patients were defined using the FDA-approved TMB cutoff of 10 mut/Mb; next, TMB-H patients were divided equally into low, moderate, and high TMB-H subgroups. TMB-H and TMB-L patients were defined using the FDA-approved TMB cutoff - 10 mut/Mb; and, TMB-H patients were further divided equally into low, moderate, and high TMB-H subgroups. Keywords: immune checkpoint blockade (ICB); tumor mutation burden (TMB); hazard ratio (HR); human cancer; overall survival (OS); cytotoxic T-lymphocyte-associated protein 4 (CTLA-4); programmed cell death protein-1 (PD-1); programmed death ligand-1 (PD-L1) EN immune checkpoint blockade (ICB) tumor mutation burden (TMB) hazard ratio (HR) human cancer overall survival (OS) cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) programmed cell death protein-1 (PD-1) programmed death ligand-1 (PD-L1) 1 5 5 06/07/22 20220603 NES 220603 Introduction Over the past decade, immunotherapy has revolutionized cancer treatment by inducing durable remission and prolonging patient survival in diverse types of cancer ([1]). [Extracted from the article]
- Published
- 2022
- Full Text
- View/download PDF
35. Dose-Dependent Effect of Tumor Mutation Burden on Cancer Prognosis Following Immune Checkpoint Blockade: Causal Implications
- Author
-
Ming Zheng
- Subjects
immune checkpoint blockade (ICB) ,tumor mutation burden (TMB) ,hazard ratio (HR) ,human cancer ,overall survival (OS) ,cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) ,Immunologic diseases. Allergy ,RC581-607 - Published
- 2022
- Full Text
- View/download PDF
36. High Grade Dermatologic Adverse Events Associated With Immune Checkpoint Blockade for Cancer
- Author
-
Alyce M. Kuo and Alina Markova
- Subjects
toxic epidermal necrolysis (TEN) ,Stevens-Johnson syndrome (SJS) ,immune checkpoint blockade (ICB) ,acute generalized exanthematous pustulosis (AGEP) ,drug reaction with eosinophilia and systemic symptoms (DRESS) ,bullous pemphigoid (BP) ,Medicine (General) ,R5-920 - Abstract
Immune checkpoint blockade (ICB) improves survival in many types of cancers including melanoma, non-small cell lung, renal cell, breast, and cervical cancers. However, many of these therapies are also associated with high grade dermatologic adverse events (DAEs), including Stevens-Johnson syndrome and toxic epidermal necrolysis (SJS/TEN), SJS/TEN-like reactions, high grade maculopapular and psoriasiform rashes, autoimmune bullous eruptions, drug reaction with eosinophilia and systemic symptoms (DRESS), and acute generalized exanthematous pustulosis (AGEP), which may limit their tolerability and use. It is important to properly identify and treat DAEs to ICB because these DAEs may be associated with positive anti-tumor response and patients may have limited options for alternative anti-cancer therapeutics. In this review, we describe high grade DAEs to increasingly used ICB agents, which target CTLA-4 and PD-1 or its ligand, PD-L1 and enable the immune system to target cancer cells. We further differentiate life-threatening adverse reactions from mimickers and report cases of serious DAEs which have been recorded in association with ICB through the FDA Adverse Events Reporting System (FAERS), which is an archive of adverse events associated with various drugs and therapeutic biologic products reported voluntarily by consumers and healthcare professionals as well as mandatorily by manufacturers. Lastly, we summarize management recommendations for these adverse events and discuss knowledge and evidence gaps in this area.
- Published
- 2022
- Full Text
- View/download PDF
37. The Chemokines Initiating and Maintaining Immune Hot Phenotype Are Prognostic in ICB of HNSCC.
- Author
-
Huang, Yuhong, Liu, Han, Liu, Xuena, Li, Nan, Bai, Han, Guo, Chenyang, Xu, Tian, Zhu, Lei, Liu, Chao, and Xiao, Jing
- Subjects
MONOCYTES ,PHENOTYPES ,IMMUNE checkpoint proteins ,ARTIFICIAL neural networks ,CHEMOKINES ,CELL death - Abstract
Background: The immune checkpoint blockade (ICB) with anti-programmed cell death protein 1(PD-1) on HNSCC is not as effective as on other tumors. In this study, we try to find out the key factors in the heterogeneous tumor-associated monocyte/macrophage (TAMM) that could regulate immune responses and predict the validity of ICB on HNSCC. Experimental Design: To explore the correlation of the TAMM heterogeneity with the immune properties and prognosis of HNSCC, we established the differentiation trajectory of TAMM by analyzing the single-cell RNA-seq data of HNSCC, by which the HNSCC patients were divided into different sub-populations. Then, we exploited the topology of the network to screen out the genes critical for immune hot phenotype of HNSCC, as well as their roles in TAMM differentiation, tumor immune cycle, and progression. Finally, these key genes were used to construct a neural net model via deep-learning framework to predict the validity of treatment with anti-PD-1/PDL-1 Results: According to the differentiation trajectory, the genes involved in TAMM differentiation were categorized into early and later groups. Then, the early group genes divided the HNSCC patients into sub-populations with more detailed immune properties. Through network topology, CXCL9, 10, 11, and CLL5 related to TAMM differentiation in the TME were identified as the key genes initiating and maintaining the immune hot phenotype in HNSCC by remarkably strengthening immune responses and infiltration. Genome wide, CASP8 mutations were found to be key to triggering immune responses in the immune hot phenotype. On the other hand, in the immune cold phenotype, the evident changes in CNV resulted in immune evasion by disrupting immune balance. Finally, based on the framework of CXCL9-11, CLL5, CD8
+ , CD4+ T cells, and Macrophage M1, the neural network model could predict the validity of PD-1/PDL-1 therapy with 75% of AUC in the test cohort. Conclusion: We concluded that the CXCL9, 10,11, and CCL5 mediated TAMM differentiation and constructed immune hot phenotype of HNSCC. Since they positively regulated immune cells and immune cycle in HNSCC, the CXCL9-11 and CCL5 could be used to predict the effects of anti-PD-1/PDL-1 therapy on HNSCC. [ABSTRACT FROM AUTHOR]- Published
- 2022
- Full Text
- View/download PDF
38. A Novel Immune-Prognosis Index Predicts the Benefit of Lung Adenocarcinoma Patients.
- Author
-
Bai, Yuquan, Pei, Yun, Xia, Liang, Ma, Lin, and Deng, Senyi
- Subjects
LUNGS ,IMMUNE checkpoint proteins ,IMMUNOLOGIC memory ,BAYESIAN analysis ,ADENOCARCINOMA ,PROGNOSIS ,DENDRITIC cells - Abstract
Background: Constructed an immune-prognosis index (IPI) and divided lung adenocarcinoma (LUAD) patients into different subgroups according to IPI score, describe the molecular and immune characteristics of patients between different IPI subgroups, and explore their response to immune checkpoint blockade (ICB) treatment. Methods: Based on the transcriptome profile of LUAD patients in TCGA and immune gene sets from ImmPort and InnateDB, 15 hub immune genes were identified through correlation and Bayesian causal network analysis. Then, IPI was constructed with 5 immune genes by using COX regression analysis and verified with external datasets (GSE30219, GSE37745, GSE68465, GSE126044 and GSE135222). Finally, the characteristics and the response to ICB treatment of LUAD patients between two different IPI subgroups were analyzed. Results: IPI was constructed based on the expression of 5 genes, including A2M, ADRB1, ADRB2, VIPR1 and PTH1R. IPI-high LUAD patients have a better overall survival than IPI-low LUAD patients, consistent with the results in the GEO cohorts. The comprehensive results showed that patients in the IPI-high subgroup were exhibited characters as metabolism-related signaling pathways activation, lower TP53 and TTN mutation rate, more infiltrations of CD8 T cells, dendritic cells and macrophages M1, especially earned more benefit from ICB treatment. In contrast, patients in the IPI-low subgroup were exhibited characters as p53 signaling pathways activation, higher TP53 and TTN mutation rate, more infiltrations of resting memory CD4 T cells, macrophages M2, immune-suppressive response and less benefit from ICB treatment. Conclusion: IPI is a potentially valuable prognostic evaluation method for LUAD, which works well in the benefit predicting of LUAD patients within ICB treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. Nanoliposome C6‐Ceramide in combination with anti‐CTLA4 antibody improves anti‐tumor immunity in hepatocellular cancer.
- Author
-
Qi, Xiaoqiang, Wu, Feng, Kim, Sung Hoon, Kaifi, Jussuf T., Kimchi, Eric T., Snyder, Helena, Illendula, Anuradha, Fox, Todd, Kester, Mark, Staveley‐O'Carroll, Kevin F., and Li, Guangfu
- Abstract
Combination therapy represents an effective therapeutic approach to overcome hepatocellular cancer (HCC) resistance to immune checkpoint blockade (ICB). Based upon previous work demonstrating that nanoliposome C6‐ceramide (LipC6) not only induces HCC apoptosis but also prevents HCC‐induced immune tolerance, we now investigate the potential of LipC6 in combination with ICB in HCC treatment. We generated orthotopic HCC‐bearing mice, which have typical features in common with human patients, and then treated them with LipC6 in combination with the antibodies (Abs) for programmed cell death protein 1 (PD‐1) or cytotoxic T‐lymphocyte antigen 4 (CTLA4). The tumor growth was monitored by magnetic resonance imaging (MRI) and the intrahepatic immune profiles were checked by flow cytometry in response to the treatments. Realtime PCR (qPCR) was used to detect the expression of target genes. The results show that LipC6 in combination with anti‐CTLA4 Ab, but not anti‐PD‐1 Ab, significantly slowed tumor growth, enhanced tumor‐infiltrating CD8+ T cells, and suppressed tumor‐resident CD4+CD25+FoxP3+ Tregs. Further molecular investigation indicates that the combinational treatment suppressed transcriptional factor Krüppel‐like Factor 2 (KLF2), forkhead box protein P3 (FoxP3), and CTLA4. Our studies suggest that LipC6 in combination with anti‐CTLA4 Ab represents a novel therapeutic approach with significant potential in activating anti‐HCC immune response and suppressing HCC growth. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
40. The Chemokines Initiating and Maintaining Immune Hot Phenotype Are Prognostic in ICB of HNSCC
- Author
-
Yuhong Huang, Han Liu, Xuena Liu, Nan Li, Han Bai, Chenyang Guo, Tian Xu, Lei Zhu, Chao Liu, and Jing Xiao
- Subjects
squamous cell carcinoma of head and neck (HNSCC) ,tumor-associated monocyte/macrophage (TAMM) ,immune checkpoint blockade (ICB) ,tumor micro-environment (TME) ,CXCL ,CCL5 ,Genetics ,QH426-470 - Abstract
Background: The immune checkpoint blockade (ICB) with anti-programmed cell death protein 1(PD-1) on HNSCC is not as effective as on other tumors. In this study, we try to find out the key factors in the heterogeneous tumor-associated monocyte/macrophage (TAMM) that could regulate immune responses and predict the validity of ICB on HNSCC.Experimental Design: To explore the correlation of the TAMM heterogeneity with the immune properties and prognosis of HNSCC, we established the differentiation trajectory of TAMM by analyzing the single-cell RNA-seq data of HNSCC, by which the HNSCC patients were divided into different sub-populations. Then, we exploited the topology of the network to screen out the genes critical for immune hot phenotype of HNSCC, as well as their roles in TAMM differentiation, tumor immune cycle, and progression. Finally, these key genes were used to construct a neural net model via deep-learning framework to predict the validity of treatment with anti-PD-1/PDL-1Results: According to the differentiation trajectory, the genes involved in TAMM differentiation were categorized into early and later groups. Then, the early group genes divided the HNSCC patients into sub-populations with more detailed immune properties. Through network topology, CXCL9, 10, 11, and CLL5 related to TAMM differentiation in the TME were identified as the key genes initiating and maintaining the immune hot phenotype in HNSCC by remarkably strengthening immune responses and infiltration. Genome wide, CASP8 mutations were found to be key to triggering immune responses in the immune hot phenotype. On the other hand, in the immune cold phenotype, the evident changes in CNV resulted in immune evasion by disrupting immune balance. Finally, based on the framework of CXCL9-11, CLL5, CD8+, CD4+ T cells, and Macrophage M1, the neural network model could predict the validity of PD-1/PDL-1 therapy with 75% of AUC in the test cohort.Conclusion: We concluded that the CXCL9, 10,11, and CCL5 mediated TAMM differentiation and constructed immune hot phenotype of HNSCC. Since they positively regulated immune cells and immune cycle in HNSCC, the CXCL9-11 and CCL5 could be used to predict the effects of anti-PD-1/PDL-1 therapy on HNSCC.
- Published
- 2022
- Full Text
- View/download PDF
41. A Novel Immune-Prognosis Index Predicts the Benefit of Lung Adenocarcinoma Patients
- Author
-
Yuquan Bai, Yun Pei, Liang Xia, Lin Ma, and Senyi Deng
- Subjects
lung adenocarcinoma ,immune cell infiltration ,immune checkpoint blockade (ICB) ,immune-prognosis index (IPI) ,network analysis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Background: Constructed an immune-prognosis index (IPI) and divided lung adenocarcinoma (LUAD) patients into different subgroups according to IPI score, describe the molecular and immune characteristics of patients between different IPI subgroups, and explore their response to immune checkpoint blockade (ICB) treatment.Methods: Based on the transcriptome profile of LUAD patients in TCGA and immune gene sets from ImmPort and InnateDB, 15 hub immune genes were identified through correlation and Bayesian causal network analysis. Then, IPI was constructed with 5 immune genes by using COX regression analysis and verified with external datasets (GSE30219, GSE37745, GSE68465, GSE126044 and GSE135222). Finally, the characteristics and the response to ICB treatment of LUAD patients between two different IPI subgroups were analyzed.Results: IPI was constructed based on the expression of 5 genes, including A2M, ADRB1, ADRB2, VIPR1 and PTH1R. IPI-high LUAD patients have a better overall survival than IPI-low LUAD patients, consistent with the results in the GEO cohorts. The comprehensive results showed that patients in the IPI-high subgroup were exhibited characters as metabolism-related signaling pathways activation, lower TP53 and TTN mutation rate, more infiltrations of CD8 T cells, dendritic cells and macrophages M1, especially earned more benefit from ICB treatment. In contrast, patients in the IPI-low subgroup were exhibited characters as p53 signaling pathways activation, higher TP53 and TTN mutation rate, more infiltrations of resting memory CD4 T cells, macrophages M2, immune-suppressive response and less benefit from ICB treatment.Conclusion: IPI is a potentially valuable prognostic evaluation method for LUAD, which works well in the benefit predicting of LUAD patients within ICB treatment.
- Published
- 2022
- Full Text
- View/download PDF
42. Harnessing epithelial-mesenchymal plasticity to boost cancer immunotherapy
- Author
-
Gu, Yuanzhuo, Zhang, Zhengkui, and ten Dijke, Peter
- Published
- 2023
- Full Text
- View/download PDF
43. Radiation Plus Anti-PD-1 Therapy for NSCLC Brain Metastases: A Retrospective Study
- Author
-
Guixiang Liao, Yuting Qian, Sumbal Arooj, Zhihong Zhao, Maosheng Yan, Zihuang Li, Hongli Yang, Tao Zheng, Gang Li, Xianming Li, and Muhammad Khan
- Subjects
non-small cell lung cancer (NSCLC) ,immunotherapy (IT) ,anti-PD-1 immunotherapy ,whole brain radiation therapy (WBRT) ,immune checkpoint blockade (ICB) ,brain metastasis (BM) ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
BackgroundRadiation therapy (RT) is the mainstay of brain metastases (BMs), and anti-PD-1 blockade has led to intracranial responses in non-small cell lung carcinoma (NSCLC) patients with BMs.ObjectiveThis study aimed to evaluate the efficacy and safety of adding anti-PD-1 blockade to RT in the management of NSCLC patients with BM in terms of survival outcome.Materials and MethodsWe retrospectively reviewed 70 NSCLC patients with BMs who were treated with whole brain radiation therapy (WBRT) between January 2016 and January 2021. Of the 70 patients, 29 additionally received anti-PD-1 therapy within 30 days of WBRT initiation. Baseline characteristics of the patients and efficacy outcomes such as progression-free survival (PFS) and overall survival (OS) were statistically compared using SPSS v26. Results were obtained using the Chi-square test/Fisher exact test, t-test, Kaplan-Meier, and Cox regression survival analyses.ResultsThe median survival for the entire cohort was 24 months (95% CI, 19.5–28.5). The median survival times for WBRT alone and WBRT plus anti-PD-1 therapy cohorts were 20 months (95% CI, 11.6–28.3) and 27 months (95% CI, 19.5–28.5), respectively (p=0.035). There was no statistical difference in PFS for the treatment cohorts (median PFS for WBRT alone: 7 months vs. 12 months for WBRT plus anti-PD-1, p=0.247). In EGFR wild-type subgroup (n=31), both PFS (p=0.037) and OS (p=0.012) were significantly improved. Only the treatment group (WBRT plus anti-PD-1) was a significant predictor of OS on univariate and multivariate analyses (p=0.040). There were no significant differences in adverse events among the treatment groups.ConclusionsNSCLC patients with BM receiving additional anti-PD-1 therapy may derive better OS than WBRT alone without any increase in adverse events. Prospective well-designed studies are warranted to validate and elucidate the additive effects of the two modalities in this group of patients.
- Published
- 2021
- Full Text
- View/download PDF
44. Radiation Plus Anti-PD-1 Therapy for NSCLC Brain Metastases: A Retrospective Study.
- Author
-
Liao, Guixiang, Qian, Yuting, Arooj, Sumbal, Zhao, Zhihong, Yan, Maosheng, Li, Zihuang, Yang, Hongli, Zheng, Tao, Li, Gang, Li, Xianming, and Khan, Muhammad
- Subjects
BRAIN metastasis ,SURVIVAL rate ,OVERALL survival ,NON-small-cell lung carcinoma ,FISHER exact test - Abstract
Background: Radiation therapy (RT) is the mainstay of brain metastases (BMs), and anti-PD-1 blockade has led to intracranial responses in non-small cell lung carcinoma (NSCLC) patients with BMs. Objective: This study aimed to evaluate the efficacy and safety of adding anti-PD-1 blockade to RT in the management of NSCLC patients with BM in terms of survival outcome. Materials and Methods: We retrospectively reviewed 70 NSCLC patients with BMs who were treated with whole brain radiation therapy (WBRT) between January 2016 and January 2021. Of the 70 patients, 29 additionally received anti-PD-1 therapy within 30 days of WBRT initiation. Baseline characteristics of the patients and efficacy outcomes such as progression-free survival (PFS) and overall survival (OS) were statistically compared using SPSS v26. Results were obtained using the Chi-square test/Fisher exact test, t-test, Kaplan-Meier, and Cox regression survival analyses. Results: The median survival for the entire cohort was 24 months (95% CI, 19.5–28.5). The median survival times for WBRT alone and WBRT plus anti-PD-1 therapy cohorts were 20 months (95% CI, 11.6–28.3) and 27 months (95% CI, 19.5–28.5), respectively (p=0.035). There was no statistical difference in PFS for the treatment cohorts (median PFS for WBRT alone: 7 months vs. 12 months for WBRT plus anti-PD-1, p=0.247). In EGFR wild-type subgroup (n=31), both PFS (p=0.037) and OS (p=0.012) were significantly improved. Only the treatment group (WBRT plus anti-PD-1) was a significant predictor of OS on univariate and multivariate analyses (p=0.040). There were no significant differences in adverse events among the treatment groups. Conclusions: NSCLC patients with BM receiving additional anti-PD-1 therapy may derive better OS than WBRT alone without any increase in adverse events. Prospective well-designed studies are warranted to validate and elucidate the additive effects of the two modalities in this group of patients. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
45. From bench to bed: the tumor immune microenvironment and current immunotherapeutic strategies for hepatocellular carcinoma
- Author
-
Yaojie Fu, Shanshan Liu, Shan Zeng, and Hong Shen
- Subjects
Hepatocellular carcinoma (HCC) ,Immunotherapy ,Oncolytic virus ,Immune checkpoint blockade (ICB) ,Adoptive cell transfer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Hepatocellular carcinoma (HCC) ranks the most common primary liver malignancy and the third leading cause of tumor-related mortality worldwide. Unfortunately, despite advances in HCC treatment, less than 40% of HCC patients are eligible for potentially curative therapies. Recently, cancer immunotherapy has emerged as one of the most promising approaches for cancer treatment. It has been proven therapeutically effective in many types of solid tumors, such as non-small cell lung cancer and melanoma. As an inflammation-associated tumor, it’s well-evidenced that the immunosuppressive microenvironment of HCC can promote immune tolerance and evasion by various mechanisms. Triggering more vigorous HCC-specific immune response represents a novel strategy for its management. Pre-clinical and clinical investigations have revealed that various immunotherapies might extend current options for needed HCC treatment. In this review, we provide the recent progress on HCC immunology from both basic and clinical perspectives, and discuss potential advances and challenges of immunotherapy in HCC.
- Published
- 2019
- Full Text
- View/download PDF
46. Pheophorbide A–Mediated Photodynamic Therapy Potentiates Checkpoint Blockade Therapy of Tumor with Low PD–L1 Expression
- Author
-
Qinli Tong, Jiaojiao Xu, Aihua Wu, Chen Zhang, Afeng Yang, Sihang Zhang, Hongzheng Lin, and Wei Lu
- Subjects
programmed death ligand 1 (PD–L1) ,pheophorbide A ,liposomes ,photodynamic therapy (PDT) ,immune checkpoint blockade (ICB) ,combined therapy ,Pharmacy and materia medica ,RS1-441 - Abstract
Although the immune checkpoint blockade (ICB) has made a great success in cancer immunotherapy, the overall response rate to the ICB, such as anti–programmed death ligand 1 (PD–L1) therapy, remains only at 20–30%. One major reason is the low expression level of the immune checkpoint in a certain type of tumor cells and its insufficient activation of the host immune system. Herein, we reported a cyclic RGD (cRGD)–modified liposomal delivery system loading the anti–PD–L1 antibody and the photosensitizer pheophorbide A (Pa), allowing a targeting of the low PD–L1 expressing 4T1 mouse breast cancer cells through the recognition of an overexpression of αvβ3 integrin on the tumor cells. The Pa–mediated photodynamic therapy (PDT) elevated the expression of PD–L1 on the tumor cells. PDT, in combination with the anti–PD–L1 therapy, promoted the activation and maturation of dendritic cells as well as the infiltration of cytotoxic T lymphocytes, resulting in the augmented antitumor immune response for the enhanced therapeutic effect. These results demonstrated the combined therapeutic effects of PDT and ICB on the tumor with low PD–L1 levels. Our study suggested that an increase in the PD–L1 expression in tumor cells by PDT would be a promising adjuvant treatment to overcome the ICB irresponsiveness.
- Published
- 2022
- Full Text
- View/download PDF
47. Soluble B7-CD28 Family Inhibitory Immune Checkpoint Proteins and Anti-Cancer Immunotherapy
- Author
-
Muhammad Khan, Sumbal Arooj, and Hua Wang
- Subjects
soluble immune checkpoints (SIC) ,alternative splice variants (ASV) ,immunotherapy (IT) ,immune checkpoint blockade (ICB) ,gene therapy ,cancer vaccination (CVax) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Co-inhibitory B7-CD28 family member proteins negatively regulate T cell responses and are extensively involved in tumor immune evasion. Blockade of classical CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) and PD-1 (programmed cell death protein-1) checkpoint pathways have become the cornerstone of anti-cancer immunotherapy. New inhibitory checkpoint proteins such as B7-H3, B7-H4, and BTLA (B and T lymphocyte attenuator) are being discovered and investigated for their potential in anti-cancer immunotherapy. In addition, soluble forms of these molecules also exist in sera of healthy individuals and elevated levels are found in chronic infections, autoimmune diseases, and cancers. Soluble forms are generated by proteolytic shedding or alternative splicing. Elevated circulating levels of these inhibitory soluble checkpoint molecules in cancer have been correlated with advance stage, metastatic status, and prognosis which underscore their broader involvement in immune regulation. In addition to their potential as biomarker, understanding their mechanism of production, biological activity, and pathological interactions may also pave the way for their clinical use as a therapeutic target. Here we review these aspects of soluble checkpoint molecules and elucidate on their potential for anti-cancer immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
48. Soluble B7-CD28 Family Inhibitory Immune Checkpoint Proteins and Anti-Cancer Immunotherapy.
- Author
-
Khan, Muhammad, Arooj, Sumbal, and Wang, Hua
- Subjects
IMMUNE checkpoint proteins ,T cells ,B cells ,IMMUNOTHERAPY ,PROGNOSIS - Abstract
Co-inhibitory B7-CD28 family member proteins negatively regulate T cell responses and are extensively involved in tumor immune evasion. Blockade of classical CTLA-4 (cytotoxic T lymphocyte-associated antigen-4) and PD-1 (programmed cell death protein-1) checkpoint pathways have become the cornerstone of anti-cancer immunotherapy. New inhibitory checkpoint proteins such as B7-H3, B7-H4, and BTLA (B and T lymphocyte attenuator) are being discovered and investigated for their potential in anti-cancer immunotherapy. In addition, soluble forms of these molecules also exist in sera of healthy individuals and elevated levels are found in chronic infections, autoimmune diseases, and cancers. Soluble forms are generated by proteolytic shedding or alternative splicing. Elevated circulating levels of these inhibitory soluble checkpoint molecules in cancer have been correlated with advance stage, metastatic status, and prognosis which underscore their broader involvement in immune regulation. In addition to their potential as biomarker, understanding their mechanism of production, biological activity, and pathological interactions may also pave the way for their clinical use as a therapeutic target. Here we review these aspects of soluble checkpoint molecules and elucidate on their potential for anti-cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
49. CU06-1004-Induced Vascular Normalization Improves Immunotherapy by Modulating Tumor Microenvironment via Cytotoxic T Cells
- Author
-
Songyi Park, Ji Hoon Oh, Dong Jin Park, Haiying Zhang, Minyoung Noh, Yeomyung Kim, Ye-Seul Kim, Hyejeong Kim, Young-Myeong Kim, Sang-Jun Ha, and Young-Guen Kwon
- Subjects
CU06-1004 ,anti-PD-1 ,combination therapy ,drug delivery ,tumor microenvironment (TME) ,immune checkpoint blockade (ICB) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Blocking the immune evasion mechanism of tumor cells has become an attractive means for treating cancers. However, the usage of a drug such as nivolumab (αPD-1), which blocks programmed cell death protein 1 (PD-1), turned out to be only effective against certain types of cancer. Especially, vascular abnormal structures of which deter delivery route by leakage and cause the poor perfusion were considered to be environment unfavorable to T cells and immune checkpoint blockade (ICB) delivery within the tumor microenvironment (TME). Herein, we report stabilization of tumor blood vessels by endothelial dysfunctional blocker CU06-1004, which modified the TME and showed synergistic effects with immunotherapy anti-PD-1 antibody. CU06-1004 combination therapy consistently prolonged the survival of tumor-bearing mice by decreasing tumor growth. T-cell infiltration increased in the tumors of the combination group, with cytotoxic CD8+ T cell activity within the tumor parenchyma upregulated compared with anti-PD-1 monotherapy. Tumor inhibition was associated with reduced hypoxia and reduced vessel density in the central region of the tumor. These effects correlated significantly with enhanced expression of IFN gamma and PD-L1 in tumors. Taken together, our findings suggest that CU06-1004 is a potential candidate drug capable of improving therapeutic efficacy of anti-PD-1 through beneficial changes in the TME.
- Published
- 2021
- Full Text
- View/download PDF
50. Combinational Immunotherapy for Hepatocellular Carcinoma: Radiotherapy, Immune Checkpoint Blockade and Beyond
- Author
-
Yun Hua Lee, David Tai, Connie Yip, Su Pin Choo, and Valerie Chew
- Subjects
radiotherapy ,immunotherapy ,immune checkpoint blockade (ICB) ,combination therapy ,hepatocellular carcinoma (HCC) ,Immunologic diseases. Allergy ,RC581-607 - Abstract
The systemic treatment landscape for advanced hepatocellular carcinoma (HCC) has experienced tremendous paradigm shift towards targeting tumor microenvironment (TME) following recent trials utilizing immune checkpoint blockade (ICB). However, limited success of ICB as monotherapy mandates the evaluation of combination strategies incorporating immunotherapy for improved clinical efficacy. Radiotherapy (RT) is an integral component in treatment of solid cancers, including HCC. Radiation mediates localized tumor killing and TME modification, thereby potentiating the action of ICB. Several preclinical and clinical studies have explored the efficacy of combining RT and ICB in HCC with promising outcomes. Greater efforts are required in discovery and understanding of novel combination strategies to maximize clinical benefit with tolerable adverse effects. This current review provides a comprehensive assessment of RT and ICB in HCC, their respective impact on TME, the rationale for their synergistic combination, as well as the current potential biomarkers available to predict clinical response. We also speculate on novel future strategies to further enhance the efficacy of this combination.
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.