19 results on '"cancer immunoediting"'
Search Results
2. Recent Advances of RNA m6A Modifications in Cancer Immunoediting and Immunotherapy
- Author
-
Ma, Shoubao, Barr, Tasha, Yu, Jianhua, Rosen, Steven T., Series Editor, Chen, Jianjun, editor, Wang, G. Greg, editor, and Lu, Jun, editor
- Published
- 2023
- Full Text
- View/download PDF
3. Cancer Immunology and the Evolution of Immunotherapy
- Author
-
Nurieva, Roza, Divenko, Margarita, Kim, Sang, Suarez-Almazor, Maria E., editor, and Calabrese, Leonard H., editor
- Published
- 2021
- Full Text
- View/download PDF
4. A tumor metastasis‐associated molecule TWIST1 is a favorable target for cancer immunotherapy due to its immunogenicity.
- Author
-
Yajima, Yuki, Kosaka, Akemi, Ishibashi, Kei, Yasuda, Shunsuke, Komatsuda, Hiroki, Nagato, Toshihiro, Oikawa, Kensuke, Kitada, Masahiro, Takekawa, Masanori, Kumai, Takumi, Ohara, Kenzo, Ohkuri, Takayuki, and Kobayashi, Hiroya
- Abstract
Although neoantigens are one of the most favorable targets in cancer immunotherapy, it is less versatile and costly to apply neoantigen‐derived cancer vaccines to patients due to individual variation. It is, therefore, important to find highly immunogenic antigens between tumor‐specific or associated antigens that are shared among patients. Considering the cancer immunoediting theory, immunogenic tumor cells cannot survive in the early phase of tumor progression including two processes: elimination and equilibrium. We hypothesized that highly immunogenic molecules are allowed to be expressed in tumor cells after an immune suppressive tumor microenvironment was established, if these molecules contribute to tumor survival. In the current study, we focused on TWIST1 as a candidate for highly immunogenic antigens because it is upregulated in tumor cells under hypoxia and promotes tumor metastasis, which is observed in the late phase of tumor progression. We demonstrated that TWIST1 had an immunogenic peptide sequence TWIST1140–162, which effectively activated TWIST1‐specific CD4+ T‐cells. In a short‐term culture system, we detected more TWIST1‐specific responses in breast cancer patients compared with in healthy donors. Vaccination with the TWIST1 peptide also showed efficient expansion of TWIST1‐reactive HTLs in humanized mice. These findings indicate that TWIST1 is a highly immunogenic shared antigen and a favorable target for cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
5. LC3B upregulation by NANOG promotes immune resistance and stem-like property through hyperactivation of EGFR signaling in immune-refractory tumor cells.
- Author
-
Kim, Suyeon, Cho, Hanbyoul, Hong, Soon-Oh, Oh, Se Jin, Lee, Hyo-Jung, Cho, Eunho, Woo, Seon Rang, Song, Joon Seon, Chung, Joon-Yong, Son, Sung Wook, Yoon, Sang Min, Jeon, Yu-Min, Jeon, Seunghyun, Yee, Cassian, Lee, Kyung-Mi, Hewitt, Stephen M., Kim, Jae-Hoon, Song, Kwon-Ho, and Kim, Tae Woo
- Subjects
CANCER immunotherapy ,PHENOTYPES ,IMMUNOPRECIPITATION ,MICROARRAY technology ,IMMUNE response - Abstract
Immune selection drives tumor cells to acquire refractory phenotypes. We previously demonstrated that cytotoxic T lymphocyte (CTL)-mediated immune pressure enriches NANOG
+ tumor cells with stem-like and immune-refractory properties that make them resistant to CTLs. Here, we report that the emergence of refractory phenotypes is highly associated with an aberrant macroautophagic/autophagic state of the NANOG+ tumor cells and that the autophagic phenotype arises through transcriptional induction of MAP1LC3B/LC3B by NANOG. Furthermore, we found that upregulation of LC3B expression contributes to an increase in EGF secretion. The subsequent hyperactivation of EGFR-AKT signaling rendered NANOG+ tumor cells resistant to CTL killing. The NANOG-LC3B-p-EGFR axis was preserved across various types of human cancer and correlated negatively with the overall survival of cervical cancer patients. Inhibition of LC3B in immune-refractory tumor models rendered tumors susceptible to adoptive T-cell transfer, as well as PDCD1/PD-1 blockade, and led to successful, long-term control of the disease. Thus, our findings demonstrate a novel link among immune-resistance, stem-like phenotypes, and LC3B-mediated autophagic secretion in immune-refractory tumor cells, and implicate the LC3B-p-EGFR axis as a central molecular target for controlling NANOG+ immune-refractory cancer. Abbreviations: ACTB: actin beta; ATG7: autophagy related 7; BafA1: bafilomycin A1 ; CASP3: caspase 3; CFSE: carboxyfluorescein succinimidyl ester; ChIP: chromatin immunoprecipitation; CI: confidence interval; CIN: cervical intraepithelial neoplasia; CSC: cancer stem cell; CTL: cytotoxic T lymphocyte; EGF: epidermal growth factor; EGFR: epidermal growth factor receptor; FIGO: International Federation of Gynecology and Obstetrics; GFP: green fluorescent protein; GZMB: granzyme B; HG-CIN: high-grade CIN; IHC: immunohistochemistry; LG-CIN: low-grade CIN; LN: lymph node; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MCL1: myeloid cell leukemia sequence 1; MLANA/MART-1: melanoma antigen recognized by T cells 1; MUT: mutant; NANOG: Nanog homeobox; PDCD1/PD-1: programmed cell death 1; PMEL/gp100: premelanosome protein; RTK: receptor tyrosine kinase; TMA: tissue microarray; WT: wild type [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
6. A stealth antigen SPESP1, which is epigenetically silenced in tumors, is a suitable target for cancer immunotherapy.
- Author
-
Kosaka, Akemi, Yajima, Yuki, Hatayama, Mayumi, Ikuta, Katsuya, Sasaki, Takaaki, Hirai, Noriko, Yasuda, Syunsuke, Nagata, Marino, Hayashi, Ryusuke, Harabuchi, Shohei, Ohara, Kenzo, Ohara, Mizuho, Kumai, Takumi, Ishibashi, Kei, Hirata‐Nozaki, Yui, Nagato, Toshihiro, Oikawa, Kensuke, Harabuchi, Yasuaki, Celis, Esteban, and Okumura, Toshikatsu
- Abstract
Recent studies have revealed that tumor cells decrease their immunogenicity by epigenetically repressing the expression of highly immunogenic antigens to survive in immunocompetent hosts. We hypothesized that these epigenetically hidden "stealth" antigens should be favorable targets for cancer immunotherapy due to their high immunogenicity. To identify these stealth antigens, we treated human lung cell line A549 with DNA methyltransferase inhibitor 5‐aza‐2′‐deoxycytidine (5Aza) and its prodrug guadecitabine for 3 d in vitro and screened it using cDNA microarray analysis. We found that the gene encoding sperm equatorial segment protein 1 (SPESP1) was re‐expressed in cell lines including solid tumors and leukemias treated with 5Aza, although SPESP1 was not detected in untreated tumor cell lines. Using normal human tissue cDNA panels, we demonstrated that SPESP1 was not detected in normal human tissue except for testis and placenta. Moreover, we found using immunohistochemistry SPESP1 re‐expression in xenografts in BALB/c‐nu/nu mice that received 5Aza treatment. To assess the antigenicity of SPESP1, we stimulated human CD4+ T‐cells with a SPESP1‐derived peptide designed using a computer algorithm. After repetitive stimulation, SPESP1‐specific helper T‐cells were obtained; these cells produced interferon‐γ against HLA‐matched tumor cell lines treated with 5Aza. We also detected SPESP1 expression in freshly collected tumor cells derived from patients with acute myeloid leukemia or lung cancer. In conclusion, SPESP1 can be classified as a stealth antigen, a molecule encoded by a gene that is epigenetically silenced in tumor cells but serves as a highly immunogenic antigen suitable for cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
7. TCR Redirected T Cells for Cancer Treatment: Achievements, Hurdles, and Goals
- Author
-
Francesco Manfredi, Beatrice Claudia Cianciotti, Alessia Potenza, Elena Tassi, Maddalena Noviello, Andrea Biondi, Fabio Ciceri, Chiara Bonini, and Eliana Ruggiero
- Subjects
TCR - T cell receptor ,genetic engineering ,cancer immunotherapy ,adoptive T cell immunotherapy ,cancer immunoediting ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Adoptive T cell therapy (ACT) is a rapidly evolving therapeutic approach designed to harness T cell specificity and function to fight diseases. Based on the evidence that T lymphocytes can mediate a potent anti-tumor response, initially ACT solely relied on the isolation, in vitro expansion, and infusion of tumor-infiltrating or circulating tumor-specific T cells. Although effective in a subset of cases, in the first ACT clinical trials several patients experienced disease progression, in some cases after temporary disease control. This evidence prompted researchers to improve ACT products by taking advantage of the continuously evolving gene engineering field and by improving manufacturing protocols, to enable the generation of effective and long-term persisting tumor-specific T cell products. Despite recent advances, several challenges, including prioritization of antigen targets, identification, and optimization of tumor-specific T cell receptors, in the development of tools enabling T cells to counteract the immunosuppressive tumor microenvironment, still need to be faced. This review aims at summarizing the major achievements, hurdles and possible solutions designed to improve the ACT efficacy and safety profile in the context of liquid and solid tumors.
- Published
- 2020
- Full Text
- View/download PDF
8. TCR Redirected T Cells for Cancer Treatment: Achievements, Hurdles, and Goals.
- Author
-
Manfredi, Francesco, Cianciotti, Beatrice Claudia, Potenza, Alessia, Tassi, Elena, Noviello, Maddalena, Biondi, Andrea, Ciceri, Fabio, Bonini, Chiara, and Ruggiero, Eliana
- Subjects
T cells ,T cell receptors ,CANCER cells ,CANCER treatment ,CELL physiology - Abstract
Adoptive T cell therapy (ACT) is a rapidly evolving therapeutic approach designed to harness T cell specificity and function to fight diseases. Based on the evidence that T lymphocytes can mediate a potent anti-tumor response, initially ACT solely relied on the isolation, in vitro expansion, and infusion of tumor-infiltrating or circulating tumor-specific T cells. Although effective in a subset of cases, in the first ACT clinical trials several patients experienced disease progression, in some cases after temporary disease control. This evidence prompted researchers to improve ACT products by taking advantage of the continuously evolving gene engineering field and by improving manufacturing protocols, to enable the generation of effective and long-term persisting tumor-specific T cell products. Despite recent advances, several challenges, including prioritization of antigen targets, identification, and optimization of tumor-specific T cell receptors, in the development of tools enabling T cells to counteract the immunosuppressive tumor microenvironment, still need to be faced. This review aims at summarizing the major achievements, hurdles and possible solutions designed to improve the ACT efficacy and safety profile in the context of liquid and solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
9. A tumor metastasis-associated molecule TWIST1 is a favorable target for cancer immunotherapy due to its immunogenicity (TWIST1特異的ヘルパーT細胞の活性化を応用したがん免疫療法に関する研究)
- Subjects
cancer immunotherapy ,tumor antigens ,cancer immunoediting ,shared ,cancer vaccine ,metastasis-associated molecules ,vaccination therapy - Abstract
Although neoantigens are one of the most favorable targets in cancer immunotherapy, it is less versatile and costly to apply neoantigen-derived cancer vaccines to patients due to individual variation. It is, therefore, important to find highly immunogenic antigens between tumor-specific or associated antigens that are shared among patients. Considering the cancer immunoediting theory, immunogenic tumor cells cannot survive in the early phase of tumor progression including two processes: elimination and equilibrium. We hypothesized that highly immunogenic molecules are allowed to be expressed in tumor cells after an immune suppressive tumor microenvironment was established, if these molecules contribute to tumor survival. In the current study, we focused on TWIST1 as a candidate for highly immunogenic antigens because it is upregulated in tumor cells under hypoxia and promotes tumor metastasis, which is observed in the late phase of tumor progression. We demonstrated that TWIST1 had an immunogenic peptide sequence TWIST1140-162 , which effectively activated TWIST1-specific CD4+ T-cells. In a short-term culture system, we detected more TWIST1-specific responses in breast cancer patients compared with in healthy donors. Vaccination with the TWIST1 peptide also showed efficient expansion of TWIST1-reactive HTLs in humanized mice. These findings indicate that TWIST1 is a highly immunogenic shared antigen and a favorable target for cancer immunotherapy.
- Published
- 2022
10. A stealth antigen SPESP1, which is epigenetically silenced in tumors, is a suitable target for cancer immunotherapy
- Author
-
Yui Hirata-Nozaki, Shohei Harabuchi, Toshihiro Nagato, Yoshinobu Ohsaki, Takaaki Sasaki, Hiroya Kobayashi, Kei Ishibashi, Toshikatsu Okumura, Yuki Yajima, Ryusuke Hayashi, Kenzo Ohara, Takumi Kumai, Syunsuke Yasuda, Esteban Celis, Kensuke Oikawa, Mayumi Hatayama, Noriko Hirai, Takayuki Ohkuri, Akemi Kosaka, Katsuya Ikuta, Mizuho Ohara, Marino Nagata, and Yasuaki Harabuchi
- Subjects
0301 basic medicine ,Cancer Research ,Antigenicity ,stealth antigens ,medicine.medical_treatment ,Epitopes, T-Lymphocyte ,Mice, Nude ,Biology ,Decitabine ,Epigenesis, Genetic ,Mice ,03 medical and health sciences ,Basic and Clinical Immunology ,0302 clinical medicine ,Antigen ,Cancer immunotherapy ,Antigens, Neoplasm ,Interferon ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Mice, Inbred BALB C ,cancer immunotherapy ,DNA methylation ,cancer immunoediting ,Immunogenicity ,Seminal Plasma Proteins ,Myeloid leukemia ,Original Articles ,T-Lymphocytes, Helper-Inducer ,General Medicine ,030104 developmental biology ,Oncology ,Cell culture ,030220 oncology & carcinogenesis ,Cancer research ,Immunohistochemistry ,Original Article ,Tumor Escape ,Immunotherapy ,Carrier Proteins ,tumor immunoescape ,medicine.drug - Abstract
Recent studies have revealed that tumor cells decrease their immunogenicity by epigenetically repressing the expression of highly immunogenic antigens to survive in immunocompetent hosts. We hypothesized that these epigenetically hidden “stealth” antigens should be favorable targets for cancer immunotherapy due to their high immunogenicity. To identify these stealth antigens, we treated human lung cell line A549 with DNA methyltransferase inhibitor 5‐aza‐2′‐deoxycytidine (5Aza) and its prodrug guadecitabine for 3 d in vitro and screened it using cDNA microarray analysis. We found that the gene encoding sperm equatorial segment protein 1 (SPESP1) was re‐expressed in cell lines including solid tumors and leukemias treated with 5Aza, although SPESP1 was not detected in untreated tumor cell lines. Using normal human tissue cDNA panels, we demonstrated that SPESP1 was not detected in normal human tissue except for testis and placenta. Moreover, we found using immunohistochemistry SPESP1 re‐expression in xenografts in BALB/c‐nu/nu mice that received 5Aza treatment. To assess the antigenicity of SPESP1, we stimulated human CD4+ T‐cells with a SPESP1‐derived peptide designed using a computer algorithm. After repetitive stimulation, SPESP1‐specific helper T‐cells were obtained; these cells produced interferon‐γ against HLA‐matched tumor cell lines treated with 5Aza. We also detected SPESP1 expression in freshly collected tumor cells derived from patients with acute myeloid leukemia or lung cancer. In conclusion, SPESP1 can be classified as a stealth antigen, a molecule encoded by a gene that is epigenetically silenced in tumor cells but serves as a highly immunogenic antigen suitable for cancer immunotherapy., We identified stealth antigen SPESP1. SPESP1 is encoded by a gene silenced in tumors epigenetically, but re‐expressed in tumors exposed to a DNA methyltransferase inhibitor. SPESP1 is a highly immunogenic because a SPESP1‐derived peptide efficiently activated CD4+ T‐cells. This suggests that SPESP1 is a favorable target for cancer immunotherapy.
- Published
- 2021
- Full Text
- View/download PDF
11. Chapter Two - The Role of Neoantigens in Naturally Occurring and Therapeutically Induced Immune Responses to Cancer.
- Author
-
Ward, Jeffrey P., Gubin, Matthew M., and Schreiber, Robert D.
- Subjects
CANCER immunology ,ANIMAL models of cancer ,COCARCINOGENS ,CANCER invasiveness ,CANCER immunotherapy ,CELL transformation - Abstract
Definitive experimental evidence from mouse cancer models and strong correlative clinical data gave rise to the Cancer Immunoediting concept that explains the dual host-protective and tumor-promoting actions of immunity on developing cancers. Tumor-specific neoantigens can serve as targets of spontaneously arising adaptive immunity to cancer and thereby determine the ultimate fate of developing tumors. Tumor-specific neoantigens can also function as optimal targets of cancer immunotherapy against established tumors. These antigens are derived from nonsynonymous mutations that occur during cellular transformation and, because they are foreign to the host genome, are not subject to central tolerance. In this review, we summarize the experimental evidence indicating that cancer neoantigens are the source of both spontaneously occurring and therapeutically induced immune responses against cancer. We also review the advances in genomics, bioinformatics, and cancer immunotherapy that have facilitated identification of neoantigens and have moved personalized cancer immunotherapies into clinical trials, with the promise of providing more specific, safer, more effective, and perhaps even more generalizable treatments to cancer patients than current immunotherapies. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
12. Novel avenues in immunotherapies for colorectal cancer.
- Author
-
Pardieck, Iris N., Jawahier, Priscilla A., Swets, Marloes, van de Velde, Cornelis J.H., and Kuppen, Peter J.K.
- Subjects
COLON cancer treatment ,CANCER immunotherapy ,CANCER immunology ,CANCER treatment ,CELLULAR immunity ,CANCER cells - Abstract
Since it is known that the immune system affects tumor growth, it has been studied if immunotherapy can be developed to combat cancer. While some successes have been claimed, the increasing knowledge on tumor-immune interactions has, however, also shown the limitations of this approach. Tumors may show selective outgrowth of cells escaped from immune control. Escape variants arise spontaneously due to the genetically instable nature of tumor cells. This is one of the most obvious limitations of cancer immunotherapy. However, new therapies are becoming available, designed to respond to tumor-immune escape. [ABSTRACT FROM PUBLISHER]
- Published
- 2016
- Full Text
- View/download PDF
13. Evolutionary Dynamics of Cancer Cell Populations under Immune Selection Pressure and Optimal Control of Chemotherapy.
- Author
-
Anita, S., Hritonenko, N., Marinoschi, G., Swierniak, A., Dimitriu, G., Lorenzi, T., and Ştefănescu, R.
- Subjects
- *
CANCER chemotherapy , *CANCER immunotherapy , *CELL populations , *EPIGENETICS , *CANCER invasiveness , *PHENOTYPES , *ANTINEOPLASTIC agents - Abstract
Increasing experimental evidence suggests that epigenetic and microenvironmental factors play a key role in cancer progression. In this respect, it is now generally recognized that the immune system can act as an additional selective pressure, which modulates tumor development and leads, through cancer immunoediting, to the selection for resistance to immune effector mechanisms. This may have serious implications for the design of effective anti-cancer protocols. Motivated by these considerations, we present a mathematical model for the dynamics of cancer and immune cells under the effects of chemotherapy and immunity-boosters. Tumor cells are modeled as a population structured by a continuous phenotypic trait, that is related to the level of resistance to receptor-induced cell death triggered by effector lymphocytes. The level of resistance can vary over time due to the effects of epigenetic modifications. In the asymptotic regime of small epimutations, we highlight the ability of the model to reproduce cancer immunoediting. In an optimal control framework, we tackle the problem of designing effective anti-cancer protocols. The results obtained suggest that chemotherapeutic drugs characterized by high cytotoxic effects can be useful for treating tumors of large size. On the other hand, less cytotoxic chemotherapy in combination with immunity-boosters can be effective against tumors of smaller size. Taken together, these results support the development of therapeutic protocols relying on combinations of less cytotoxic agents and immune-boosters to fight cancer in the early stages. [ABSTRACT FROM PUBLISHER]
- Published
- 2014
- Full Text
- View/download PDF
14. Cancer immunoediting: antigens, mechanisms, and implications to cancer immunotherapy.
- Author
-
Vesely, Matthew D. and Schreiber, Robert D.
- Subjects
- *
CANCER immunotherapy , *ANTIGENS , *CANCER cells , *TUMOR immunology , *INFLAMMATION , *ANTINEOPLASTIC agents , *CANCER genetics - Abstract
Accumulated data from animal models and human cancer patients strongly support the concept that the immune system can identify and control nascent tumor cells in a process called cancer immunosurveillance. In addition, the immune system can also promote tumor progression through chronic inflammation, immunoselection of poorly immunogenic variants, and suppressing antitumor immunity. Together, the dual host-protective and tumor-promoting actions of immunity are referred to as cancer immunoediting. The current framework of cancer immunoediting is a dynamic process comprised of three distinct phases: elimination, equilibrium, and escape. Recently, we demonstrated that immunoselection by CD8+ T cells of tumor variants lacking strong tumor-specific antigens represents one mechanism by which cancer cells escape tumor immunity and points toward the future of personalized cancer therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2013
- Full Text
- View/download PDF
15. Of Mice, Dogs, Pigs, and Men: Choosing the Appropriate Model for Immuno-Oncology Research
- Author
-
Daniel R. Principe, Gregers Jungersen, Kyle M. Schachtschneider, Nana Haahr Overgaard, Timothy M. Fan, and Lawrence B. Schook
- Subjects
0301 basic medicine ,Cancer immunoediting ,Swine ,040301 veterinary sciences ,medicine.medical_treatment ,Translational immunology ,Medical Oncology ,General Biochemistry, Genetics and Molecular Biology ,Translational Research, Biomedical ,0403 veterinary science ,Mice ,03 medical and health sciences ,Dogs ,Immune system ,SDG 3 - Good Health and Well-being ,Cancer immunotherapy ,Neoplasms ,medicine ,Animals ,Humans ,Porcine cancer models ,Comparative oncology ,business.industry ,Immunogenicity ,Cancer ,04 agricultural and veterinary sciences ,General Medicine ,Immunotherapy ,medicine.disease ,Genetically modified organism ,Immunosurveillance ,Disease Models, Animal ,030104 developmental biology ,Immunoediting ,Cancer research ,Animal Science and Zoology ,Canine cancer models ,business - Abstract
The immune system plays dual roles in response to cancer. The host immune system protects against tumor formation via immunosurveillance; however, recognition of the tumor by immune cells also induces sculpting mechanisms leading to a Darwinian selection of tumor cell variants with reduced immunogenicity. Cancer immunoediting is the concept used to describe the complex interplay between tumor cells and the immune system. This concept, commonly referred to as the three E’s, is encompassed by 3 distinct phases of elimination, equilibrium, and escape. Despite impressive results in the clinic, cancer immunotherapy still has room for improvement as many patients remain unresponsive to therapy. Moreover, many of the preclinical results obtained in the widely used mouse models of cancer are lost in translation to human patients.To improve the success rate of immuno-oncology research and preclinical testing of immune-based anticancer therapies, using alternative animal models more closely related to humans is a promising approach. Here, we describe 2 of the major alternative model systems: canine (spontaneous) and porcine (experimental) cancer models. Although dogs display a high rate of spontaneous tumor formation, an increased number of genetically modified porcine models exist. We suggest that the optimal immuno-oncology model may depend on the stage of cancer immunoediting in question. In particular, the spontaneous canine tumor models provide a unique platform for evaluating therapies aimed at the escape phase of cancer, while genetically engineered swine allow for elucidation of tumor-immune cell interactions especially during the phases of elimination and equilibrium.
- Published
- 2018
- Full Text
- View/download PDF
16. Immune checkpoint blockade opens an avenue of cancer immunotherapy with a potent clinical efficacy
- Author
-
Keishi Adachi and Koji Tamada
- Subjects
Cancer Research ,medicine.medical_treatment ,chemical and pharmacologic phenomena ,Ipilimumab ,Pembrolizumab ,Biology ,Immune system ,Cancer immunotherapy ,Neoplasms ,PD-1/PDL-1 pathway ,medicine ,Animals ,Humans ,Review Articles ,cancer immunoediting ,General Medicine ,Immunotherapy ,Immune checkpoint ,Anti-PD-L1 Ab ,cancer immunosurveillance ,immune checkpoint molecules ,Oncology ,Tumor Escape ,Immunology ,Nivolumab ,medicine.drug - Abstract
Recent progress in tumor immunology has revealed that tumors generate immunologically restrained milieu during the process of their growth, which facilitates the escape of tumors from host immune systems. Immune checkpoint molecules, which transduce co-inhibitory signals to immuno-competent cells, are one of the most important components conferring the immunosuppressive capacity in the tumor microenvironment. Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death-1 (PD-1) are typical immune checkpoint molecules intimately involved in the suppression of anti-tumor immunity. Antibodies against those molecules have been developed, such as ipilimumab (anti-CTLA-4 antibody), nivolumab and pembrolizumab (anti-PD-1 antibody), and have been approved by regulatory agencies and used in some countries. Treatment with these antibodies demonstrates previously unobserved clinical efficacies superior to the conventional therapies. In this review, we first discuss the escape mechanisms of cancer from host immune systems, and then focus on the recent advances in immune checkpoint blockade therapy and on the new findings of related immune reactions, aiming to provide a better understanding of the novel cancer immunotherapies.
- Published
- 2015
- Full Text
- View/download PDF
17. Novel avenues in immunotherapies for colorectal cancer
- Author
-
Marloes Swets, Cornelis J.H. van de Velde, Peter J. K. Kuppen, Iris N. Pardieck, and Priscilla A. Jawahier
- Subjects
0301 basic medicine ,Colorectal cancer ,medicine.medical_treatment ,colorectal cancer ,Immune control ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Cancer immunotherapy ,medicine ,Animals ,Humans ,tumor microenvironment ,Genetic Predisposition to Disease ,Tumor growth ,Tumor microenvironment ,clinical trials ,Hepatology ,business.industry ,cancer immunoediting ,Macrophages ,Gastroenterology ,Cancer ,Immunotherapy ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,CRC ,immune system ,Phenotype ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Tumor Escape ,immunotherapy ,Colorectal Neoplasms ,business - Abstract
Since it is known that the immune system affects tumor growth, it has been studied if immunotherapy can be developed to combat cancer. While some successes have been claimed, the increasing knowledge on tumor-immune interactions has, however, also shown the limitations of this approach. Tumors may show selective outgrowth of cells escaped from immune control. Escape variants arise spontaneously due to the genetically instable nature of tumor cells. This is one of the most obvious limitations of cancer immunotherapy. However, new therapies are becoming available, designed to respond to tumor-immune escape.
- Published
- 2016
18. Mechanisms of specific immune response interactions with tumor cells
- Author
-
Kaššák, Filip, Hořejší, Václav, and Černý, Jan
- Subjects
tumor-specific antigens ,cancer immunoediting ,nádorová imunita ,PD-1 ,blokáda imunitních "kontolních bodů" ,tumor-associated antigens ,imunoeditace ,imunoterapie nádorů ,immunosurveillance ,s tumorem asociované antigeny ,cancer immunotherapy ,imunitní dohled ,TGF-β ,immune checkpoint blockade ,cancer immunity ,NKG2D ,tumor-specifické antigeny ,CTLA-4 - Abstract
Interactions between the immune system and tumors have been among the highlights of present immunological research. An extensive body of new knowledge recently substantiated the long-presumed concept of cancer immunosurveillance. Immune system searches the organism for cells expressing tumor antigens or cellular stress signals and destroys them. T-cells, NK-cells and dendritic cells, as well as cytokine signaling and direct cell cytotoxicity play dominant role in this process. However, a fraction of nascent tumors can evade these mechanisms and create a dynamic equilibrium, gradually sculpting its phenotype by clonal selection. Eventually, tumor cells escape immune control by concealing themselves from recognition or by actively subjugating local immune response. This immunosubversion results in formation of immunosuppressive tumor microenvironment by recruiting protumorigenic cell populations, such as Treg cells, macrophages and myeloid derived suppressor cells. Soluble signaling molecules, as well as surface- expressed immune checkpoint molecules are exploited by tumor cells for inhibition of anti-tumor immunity. Highly effective therapeutic antibodies blocking these checkpoints have been developed for clinical use, with many more in current trials. Several other promising immunotherapeutic...
- Published
- 2016
19. Tumor suppressor maspin as a modulator of host immune response to cancer
- Author
-
Wael Sakr, Sijana H. Dzinic, Shijie Sheng, Marian Wahba, Daniel S. M. Oliveira, and M. Margarida Bernardo
- Subjects
Anticancer immunity ,medicine.medical_treatment ,Review Article ,Biology ,immunogenicity ,Immune system ,Cancer immunotherapy ,Cancer stem cell ,Neoplasms ,medicine ,Animals ,Humans ,innate immunity ,Serpins ,lcsh:R5-920 ,Innate immune system ,immunosuppression ,cancer immunoediting ,Maspin ,Immunotherapy ,adaptive immunity ,Acquired immune system ,Immunosurveillance ,Immunology ,lcsh:Medicine (General) - Abstract
Despite the promising clinical outcome, the primary challenge of the curative cancer immunotherapy is to overcome the dichotomy of the immune response: tumor-evoked immunostimulatory versus tumor-induced immunosuppressive. The goal needs to be two-fold, to re-establish sustainable antitumor-cancer immunity and to eliminate immunosuppression. The successful elimination of cancer cells by immunosurveillance requires the antigenic presentation of the tumor cells or tumor-associated antigens and the expression of immunostimulatory cytokines and chemokines by cancer and immune cells. Tumors are heterogeneous and as such, some of the tumor cells are thought to have stem cell characteristics that enable them to suppress or desensitize the host immunity due to acquired epigenetic changes. A central mechanism underlying tumor epigenetic instability is the increased histone deacetylase (HDAC)-mediated repression of HDAC-target genes regulating homeostasis and differentiation. It was noted that pharmacological HDAC inhibitors are not effective in eliminating tumor cells partly because they may induce immunosuppression. We have shown that epithelial-specific tumor suppressor maspin, an ovalbumin-like non-inhibitory serine protease inhibitor, reprograms tumor cells toward better differentiated phenotypes by inhibiting HDAC1. Recently, we uncovered a novel function of maspin in directing host immunity towards tumor elimination. In this review, we discuss the maspin and maspin/HDAC1 interplay in tumor biology and immunology. We propose that maspin based therapies may eradicate cancer.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.