• Title/Summary/Keyword: Anti-cancer immunity

Search Result 94, Processing Time 0.024 seconds

A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity

  • Kim, KyeongJin;Yang, Wen-Hao;Jung, Youn-Sang;Cha, Jong-ho
    • BMB Reports
    • /
    • v.53 no.10
    • /
    • pp.512-520
    • /
    • 2020
  • T-cell-based cancer immunotherapies, such as immune checkpoint blockers (ICBs) and chimeric antigen receptor (CAR)-T-cells, have significant anti-tumor effects against certain types of cancer, providing a new paradigm for cancer treatment. However, the activity of tumor infiltrating T-cells (TILs) can be effectively neutralized in the tumor microenvironment (TME) of most solid tumors, rich in various immunosuppressive factors and cells. Therefore, to improve the clinical outcomes of established T-cell-based immunotherapy, adjuvants that can comprehensively relieve multiple immunosuppressive mechanisms of TME are needed. In this regard, recent studies have revealed that metformin has several beneficial effects on anti-tumor immunity. In this mini-review, we understand the immunosuppressive properties of TME and how metformin comprehensively enhances anti-tumor immunity. Finally, we will discuss this old friend's potential as an adjuvant for cancer immunotherapy.

Acetate decreases PVR/CD155 expression via PI3K/AKT pathway in cancer cells

  • Tran, Na Ly;Lee, In Kyu;Choi, Jungkyun;Kim, Sang-Heon;Oh, Seung Ja
    • BMB Reports
    • /
    • v.54 no.8
    • /
    • pp.431-436
    • /
    • 2021
  • In recent years, restoring anti-tumor immunity has garnered a growing interest in cancer treatment. As potential therapeutics, immune checkpoint inhibitors have demonstrated benefits in many clinical studies. Although various methods have been applied to suppress immune checkpoints to boost anti-tumor immunity, including the use of immune checkpoint inhibitors, there are still unmet clinical needs to improve the response rate of cancer treatment. Here, we show that acetate can suppress the expression of poliovirus receptor (PVR/CD155), a ligand for immune checkpoint, in colon cancer cells. We demonstrated that acetate treatment could enhance effector responses of CD8+ T cells by decreasing the expression of PVR/CD155 in cancer cells. We also found that acetate could reduce the expression of PVR/CD155 by deactivating the PI3K/AKT pathway. These results demonstrate that acetate-mediated expression of PVR/CD155 in cancer cells might potentiate the anti-tumor immunity in the microenvironment of cancer. Our findings indicate that maintaining particular acetate concentrations could be a complementary strategy in current cancer treatment.

The Anti-tumor Activity of Vitamin C via the Increase of Fas (CD95) and MHC I expression on Human Stomach Cancer Cell Line, SNU1

  • Yu, Yeon-Sil;Bae, Se-Yeon;Kim, Hye-Min;Kim, Ye-Jin;Chu, Nag-Bum;Chu, Nag-Kyun;Kang, Jae-Seung;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.11 no.4
    • /
    • pp.210-215
    • /
    • 2011
  • It is already known that high concentration of vitamin C induces apoptosis on tumor cells. However, there is no report regarding the function of vitamin C on the modulation of immune susceptibility of cancer. Therefore, we investigated whether vitamin C can modulate immune susceptibility of tumor cells, especially on the induction of Fas-mediated apoptosis. First, the optimal concentration of vitamin C, which cannot induce damages on tumor cells for 36 hrs. We found that 2 mM of vitamin C did not show harmful effect. In addition, the optimal concentration of agonistic anti-Fas Abs for 18 hrs was examined. As a result, 400 ng/ml of agonistic anti-Fas Abs did not induce apoptosis on tumor cells. Next, we tried to find the effect of 2 mM of vitamin C on the modulation of the susceptibility to agonistic anti-Fas Abs. When tumor cells were cultured with 400 ng/ml of agonistic anti-Fas Abs for 18 hrs, after pre-treatment with 2 mM of vitamin C for 24 hrs, viability of cells was decreased. Interestingly, we found that the expression of Fas (CD95) and MHC class I was increased by the treatment of vitamin C. Taken together, vitamin C increases the susceptibility of tumor cells to anti-Fas Abs and the expression of Fas (CD95) and MHC class I on tumor cells.

The CD28-B7 Family in Anti-Tumor Immunity: Emerging Concepts in Cancer Immunotherapy

  • Leung, Joanne;Suh, Woong-Kyung
    • IMMUNE NETWORK
    • /
    • v.14 no.6
    • /
    • pp.265-276
    • /
    • 2014
  • The interactions between B7 molecules and CD28-family receptors are crucial in the regulation of adaptive cellular immunity. In cancer, the aberrant expression of co-inhibitory B7 molecules has been attributed to reduced anti-tumor immunity and cancer immune evasion, prompting the development of cancer therapeutics that can restore T cell function. Murine tumor models have provided significant support for the targeting of multiple immune checkpoints involving CTLA-4, PD-1, ICOS, B7-H3 and B7-H4 during tumor growth, and clinical studies investigating the therapeutic effects of CTLA-4 and PD-1 blockade have shown exceptionally promising results in patients with advanced melanoma and other cancers. The expression pattern of co-inhibitory B7 ligands in the tumor microenvironment has also been largely correlated with poor patient prognosis, and recent evidence suggests that the presence of several B7 molecules may predict the responsiveness of immunotherapies that rely on pre-existing tumor-associated immune responses. While monotherapies blocking T cell co-inhibition have beneficial effects in reducing tumor burden, combinatorial immunotherapy targeting multiple immune checkpoints involved in various stages of the anti-tumor response has led to the most substantial impact on tumor reduction. In this review, we will examine the contributions of B7- and CD28-family members in the context of cancer development, and discuss the implications of current human findings in cancer immunotherapy.

Evaluation of Anti-tumor Immunity in Cancer Patients through Peripheral Blood Natural Killer Cell Measurements: A Review (말초혈액 내 NK cell 측정을 통한 암환자의 종양 면역 기능 평가 가능성 고찰)

  • Jang, Sung-Il;Choi, Jung-Eun;Lee, Yeon-Weol;Cho, Chong-Kwan;Yoo, Hwa-Seung
    • Journal of Haehwa Medicine
    • /
    • v.21 no.1
    • /
    • pp.87-96
    • /
    • 2012
  • Aim : Natural killer (NK) cells are cytotoxic lymphocytes that lyse certain tumor- and virus-infected cells without any prior stimulation or immunization. This article aims to review the significance of evaluating peripheral blood NK cells to predict anti-tumor immune function and prognosis in cancer patients. Methods : PubMed was used to create a database for this review. Search words of cancer, natural killer cell, prognosis were used to retrieve related articles. References of the collected articles were also reviewed. Results : Current evidence indicates that decreased or absent NK cell count or activity is mostly associated with the development or progression of cancer. In patients with various types of cancer, NK cell activity was mostly associated with the cancer prognosis and survival despite some conflicting results. Conclusion : The data shows the evaluation of anti-tumor immunity in cancer patients through natural killer cell measurement still remains a controversial matter. However, it is clear that the NK cell activity plays an important role in cancer and is associated with prevention of both early stage and metastatic cancer.

Nanotechnology Meet Immunology: Nanomaterials for Enhanced Immunity

  • Im, Yong-Taek
    • Proceedings of the Korean Vacuum Society Conference
    • /
    • 2013.08a
    • /
    • pp.92.2-92.2
    • /
    • 2013
  • The design and chemical synthesis of multifunctional nanomaterials have been providing potential applications in biomedical fields such as molecular imaging and drug delivery. Recently, bio-derived and/or synthetic nanostructured materials capable of modulating the immune system have been also issues of interest in immunology-related nanomedicine fields. In this talk, the recent research results on the development of nanostructured materials for enhanced immunity would be presented. I will introduce the chemical strategy for the combination of nanostructured materials and bioactive compounds to improve both anti-cancer immunity and vaccine delivery efficiency.

  • PDF

Identification of CM1 as a Pathogenic Factor in Inflammatory Diseases and Cancer

  • Bae, Se-Yeon;Kim, Hyem-In;Yu, Yeon-Sil;Lee, Na-Eun;Kong, Joo-Myoung;Kim, Hang-Rae;Hwang, Young-Il;Song, Yeong-Wook;Kang, Jae-Seung;Lee, Wang-Jae
    • IMMUNE NETWORK
    • /
    • v.11 no.3
    • /
    • pp.175-181
    • /
    • 2011
  • Background: CM1 (centrocyte/-blast marker 1) was defined by a mAb against concavabalin-A (ConA) activated PBMC. It is expressed in germinal center of human tonsil and on the surface of activated PBMC as well as cancer cells. Recently, increased productions of pro-inflammatory mediators were detected from activated PBMC by CM1 ligation. Methods: However, there is a limitation to explain the exact role of CM1 on inflammation and its related mechanisms, since the identity of CM1 is still not clarified. In our previous study, we have already confirmed that soluble form of CM1 was produced by Raji. Therefore, we performed Q-TOF analysis after immunoprecipitation of concentrated Raji culture supernatant using anti-CM1 mAbs. Results: As a result, we found that CM1 is identical to enolase-1(ENO1), a glycolytic enzyme, and we confirmed that results by silencing ENO1 using siRNA. It was also confirmed through competition assay between anti-CM1 and anti-ENO1 mAbs. Finally, we investigated the possible role of CM1 in inflammatory response and cancer. The ligation of CM1 on Raji cells with anti-CM1 mAbs induces the extensive production of prostaglandin $E_2(PGE_2)$. In addition, the increased activity of matrix metalloproteinase (MMP)-2/9 was shown in NCI-N87, stomach cancer cell line by CM1 stimulation. Conclusion: CM1 is identical to ENO1 and it might be an important role in the regulation of inflammatory responses.

A non-replicating oncolytic vector as a novel therapeutic tool against cancer

  • Kaneda, Yasufumi
    • BMB Reports
    • /
    • v.43 no.12
    • /
    • pp.773-780
    • /
    • 2010
  • Cancers are still difficult targets despite recent advances in cancer therapy. Due to the heterogeneity of cancer, a single-treatment modality is insufficient for the complete elimination of cancer cells. Therapeutic strategies from various aspects are needed. Gene therapy has been expected to bring a breakthrough to cancer therapy, but it has not yet been successful. Gene therapy also should be combined with other treatments to enhance multiple therapeutic pathways. In this view, gene delivery vector itself should be equipped with intrinsic anti-cancer activities. HVJ (hemagglutinating virus of Japan; Sendai virus) envelope vector (HVJ-E) was developed to deliver therapeutic molecules. HVJ-E itself possessed anti-tumor activities such as the generation of anti-tumor immunities and the induction of cancer-selective apoptosis. In addition to the intrinsic anti-tumor activities, therapeutic molecules incorporated into HVJ-E enabled to achieve multi-modal therapeutic strategies in cancer treatment. Tumor-targeting HVJ-E was also developed. Thus, HVJ-E will be a novel promising tool for cancer treatment.

CD4+ cytotoxic T cells: an emerging effector arm of anti-tumor immunity

  • Seongmin Jeong;Nawon Jang;Minchae Kim;Il-Kyu Choi
    • BMB Reports
    • /
    • v.56 no.3
    • /
    • pp.140-144
    • /
    • 2023
  • While CD8+ cytotoxic T cells have long been considered the primary effector in controlling tumors, the involvement of CD4+ "helper" T cells in anti-tumor immunity has been underappreciated. The investigations of intra-tumoral T cells, fueled by the recent advances in genomic technologies, have led to a rethinking of the indirect role of CD4+ T cells that have traditionally been described as a "helper". Accumulating evidence from preclinical and clinical studies indicates that CD4+ T cells can acquire intrinsic cytotoxic properties and directly kill various types of tumor cells in a major histocompatibility complex class II (MHC-II)-dependent manner, as opposed to the indirect "helper" function, thus underscoring a potentially critical contribution of CD4+ cytotoxic T cells to immune responses against a wide range of tumor types. Here, we discuss the biological properties of anti-tumor CD4+ T cells with cytotoxic capability and highlight the emerging observations suggesting their more significant role in anti-tumor immunity than previously appreciated.