scholarly journals Bifidobacterium Strain-Specific Enhances the Efficacy of Cancer Therapeutics in Tumor-Bearing Mice

Cancers ◽  
2021 ◽  
Vol 13 (5) ◽  
pp. 957
Author(s):  
Youngmin Yoon ◽  
Gihyeon Kim ◽  
Bu-Nam Jeon ◽  
Sungsoon Fang ◽  
Hansoo Park

Colorectal cancer (CRC) is among the leading causes of cancer-related death in the world. The development of CRC is associated with smoking, diet, and microbial exposure. Previous studies have shown that dysbiosis of the gut microbiome affects cancer development, because it leads to inflammation and genotoxicity. Supplementation with specific microbiota induces anti-tumor effects by enhancing of anti-tumor immunity. Here, we observed that supplementation with either of two B. breve strains reduces tumor growth in MC38 colon carcinoma-bearing mice. Interestingly, only one B. breve strain boosted the efficacy of cancer therapeutics, including oxaliplatin and PD-1 blockade. Extensive immune profiling and transcriptomic analysis revealed that the boosting B. breve strain augments lymphocyte-mediated anti-cancer immunity. Our results suggest that supplementation with B. breve strains could potentially be used as a strategy to enhance the efficacy of CRC therapeutics.

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A717-A717
Author(s):  
Abigail Overacre-Delgoffe ◽  
Anthony Cillo ◽  
Hannah Bumgarner ◽  
Ansen Burr ◽  
Justin Tometich ◽  
...  

BackgroundColorectal cancer remains one of the most common and deadliest cancers worldwide and effective therapies are lacking. While immunotherapy has revolutionized treatment for many cancers, the overwhelming majority of colorectal cancer patients are non-responsive and the 5-year survival rate for advanced disease is <20%. Immunotherapeutic response has been associated with select members of the microbiome in melanoma; however, the potential benefit in colorectal cancer and the underlying mechanisms remain unclear. We sought to determine how specific members of the intestinal microbiome affect anti-tumor immunity in colorectal cancer (CRC) in hopes of discovering novel treatments and revealing potential hurdles to current therapeutic response in CRC patients.MethodsWe utilized a carcinogen-induced mouse model of CRC and colonized half of the tumor-bearing mice with Helicobacter hepaticus (Hhep) 7 weeks post AOM. Tumor number was assessed 12 weeks post AOM. We isolated lymphocytes from the lamina propria, colonic epithelium, mesenteric lymph nodes, and tumor(s) to track the spatial and transcriptional Hhep-specific and endogenous immune responses during tumor progression through 5’ single cell RNAseq, flow cytometry, and immunofluorescence. In addition, we utilized 16S sequencing and FISH to track Hhep colonization, location within the colon, and its impact on the surrounding microbiome.ResultsWe have found that rational modification of the microbiome of colon tumor-bearing mice through addition of a single bacteria, Hhep, led to tumor control or clearance and a significant survival advantage. Colonization led to the expansion of the lymphatic network and development of numerous peri- or intra-tumoral tertiary lymphoid structures (TLS) composed of Hhep-specific CD4 T follicular helper cells (TFH) as well as the bacteria itself. This led to an overall ‘heating’ of the tumor, wherein we saw an increase of CD4 T cell infiltration to the tumor core as well as an increase in CD103+ type 1 DC (cDC1) recruitment through increased chemokines such as CCL5 and XCL1. Hhep-specific TFH were both necessary and sufficient to drive TLS formation, increased immune invasion, and anti-tumor immunity.ConclusionsWe have shown that addition of a single bacteria, Hhep, leads to a reduction in CRC tumor burden or clearance through lymphatic expansion, TLS formation, and remodeling of the tumor microenvironment, and that Hhep-specific T cells are required for tumor control. These studies suggest that rational modification of the microbiome and microbiome-specific T cells can positively impact anti-tumor immunity and may represent a unique immunotherapeutic target to turn resistant tumors into responsive tumors.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A880-A880
Author(s):  
Abigail Overacre-Delgoffe ◽  
Hannah Bumgarner ◽  
Anthony Cillo ◽  
Ansen Burr ◽  
Justin Tometich ◽  
...  

BackgroundColorectal cancer (CRC) is one of the most common and deadly cancers in the US, and the survival rate for advanced cases is poor. While immunotherapy has revolutionized cancer treatment, CRC remains largely unresponsive, with only ~6% of patients responding to anti-PD1. Specific microbiome signatures are associated with anti-PD1 response in melanoma patients; however, the underlying mechanism remains unclear. While the microbiome in cancer patients has been extensively studied, the endogenous immune response to these microbes and the subsequent effects on cancer immunity remain unstudied. Most microbes reside within the gut, and bacteria that adhere to the intestinal epithelium can stimulate bacteria-specific immune responses. Therefore, we hypothesized that the microbiome, especially adherent, immunogenic bacteria, may support anti-tumor immunity through activation of local microbiota-specific T cells.MethodsUsing a carcinogen-induced mouse model of CRC, we sought to determine the impact of microbiome modulation on the anti-tumor immune response. We colonized tumor-bearing mice with Helicobacter hepaticus (Hhep) and assessed tumor burden, survival, and immune infiltration. Lymphocytes were isolated from the tumor and surrounding tissue when tumors were terminal (12 weeks). We utilized TCR transgenic mice and MHC class II tetramers to track the spatial and transcriptional Hhep-specific T cell response through 5’ single cell RNAseq, flow cytometry, and spectral immunofluorescence.ResultsHhep colonization in tumor-bearing mice led to decreased tumor burden and significantly improved survival. Interestingly, colonization induced activation of Hhep-specific T follicular helper cells (TFHs) that supported formation of mature peri- or intra-tumoral tertiary lymphoid structures (TLS). The presence of TLS led to increased infiltration of cytotoxic lymphocytes (T and NK cells) within the tumor core. Surprisingly, the anti-tumor response was dependent on CD4+ T and B cells but not CD8+ T cells. Using TFH KO mice, we found that Hhep-specific CD4+ T cells were both necessary and sufficient to drive TLS maturation and anti-tumor immunity.ConclusionsHere, we demonstrate that addition of a single bacterial species after tumor formation leads to a reduction in CRC tumor burden and increased survival through TLS maturation. This microbiome-dependent remodeling of the tumor microenvironment is driven by Hhep-specific TFH cells that are both necessary and sufficient for tumor control, demonstrating for the first time that microbiota-specific T cells contribute to anti-tumor immunity. Overall, these findings suggest that microbiome modulation and the subsequent microbiota-specific CD4+ T cell response may represent a new variety of immunotherapies for cancers that remain resistant to checkpoint blockade.


2020 ◽  
Vol 21 (15) ◽  
pp. 5353 ◽  
Author(s):  
Hsiuying Wang

Colorectal cancer (CRC) is the third leading cause of cancer death in the world, and its incidence is rising in developing countries. Treatment with 5-Fluorouracil (5-FU) is known to improve survival in CRC patients. Most anti-cancer therapies trigger apoptosis induction to eliminate malignant cells. However, de-regulated apoptotic signaling allows cancer cells to escape this signaling, leading to therapeutic resistance. Treatment resistance is a major challenge in the development of effective therapies. The microRNAs (miRNAs) play important roles in CRC treatment resistance and CRC progression and apoptosis. This review discusses the role of miRNAs in contributing to the promotion or inhibition of apoptosis in CRC and the role of miRNAs in modulating treatment resistance in CRC cells.


2018 ◽  
Vol 2 (S1) ◽  
pp. 15-16
Author(s):  
Jonathan B. Mitchem ◽  
Yue Guan ◽  
Mark Daniels ◽  
Emma Teixeiro

OBJECTIVES/SPECIFIC AIMS: Despite significant advances in screening and treatment, colorectal cancer is the second leading cancer killer in the United States today. Some of the most promising recent developments in cancer therapy have come from immune-based therapy. Immune-based therapy, however, has shown limited utility in patients with colorectal cancer. Studies have previously shown that certain chemotherapy regimens may be more effective in combination with immune-based therapy due to induction of inflammation in the tumor microenvironment. In this study, we sought to determine how standard chemotherapy (FOLFOX) affects the generation of antigen-specific anti-tumor immunity in colorectal cancer. METHODS/STUDY POPULATION: To determine the how antigen-specific immunity and T cell responses are affected by FOLFOX, we utilized a model antigen expressing murine colon cancer cell line syngeneic to C57BL/6 (MC38-CEA). Treatment was initiated when tumor size reached 50 mm2. Mice were treated with either vehicle (PBS), 5-Fluorouracil (5-FU), Oxaliplatin, or combination (FOLFOX). Antigen-specific cytotoxic T cell (tet+Tc) were detected using Db-CEA-tetramer obtained from the NIH-tetramer core facility. Flow cytometry was performed for phenotypic analysis and tetramer positivity. Tumor growth was measured using standard caliper measurements. Statistical analysis was performed using t-test for continuous variables and ANOVA was used when comparing multiple groups. Statistical analysis was performed using SPSS. All arms were completed with n=3–7. RESULTS/ANTICIPATED RESULTS: To determine how systemic treatment with chemotherapy affects cytotoxic T cell development (Tc), we established that we could detect antigen-specific Tc (tet+Tc) in the spleen, tumor, and draining lymph nodes of tumor-bearing mice. After establishing that the system worked appropriately, tumor-bearing mice were treated with different chemotherapy regimens and tumor growth was monitored. As expected, the combination of FOLFOX was significantly better than either drug individually (2-way ANOVA, p<0.01). FOLFOX therapy also showed a significant (p<0.05) increase in the number of tumor-associated tet+Tc, and tet+Tc expressing phenotypic markers of effector (Te) and resident memory (Trm) subsets. Tumor-associated tet+Tc highly expressed PD-1 (>50%); however, this was not significantly different between treatment or vehicle arms. Since 5-FU, one component of FOLFOX has previously shown a selective reduction of myeloid-derived suppressor cells, we also investigated the myeloid compartment. There were no significant differences in conventional or plasmacytoid dendritic cells, myeloid-derived suppressor cells, or tumor-associated macrophages. DISCUSSION/SIGNIFICANCE OF IMPACT: The future of cancer care involves multi-modality care tailored to patients. To more effectively combine therapy it is critical that we understand how currently utilized therapy works. In this study, we show that the primary chemotherapy regimen utilized in colorectal cancer increases tumor-associated antigen-specific cytotoxic T cells and the majority of these cells are PD-1 positive. This suggests that FOLFOX may work in concert with immune-based therapy when selected appropriately. Further study is warranted to determine optimal combination therapy and ways to maximize anti-tumor immunity in order to improve the treatment of patients with this deadly disease.


Author(s):  
A. M. Karamzin ◽  
A. V. Ropot ◽  
R. E. Boshian

Colorectal cancer is a disease that is far from the last place in the morbidity statistics in the Russian Federation and in the world. Along with well-known risk factors for the development of this pathology, some representatives of the intestinal microbiota are possible to participate in this process. Some studies suggest that Akkermansia muciniphila, a mucin-degrading bacterium, is associated with colorectal cancer development, but other studies cast doubt on this statement. In this review, we describe a series of studies devoted to determining the dependence of colorectal cancer on the amount of A. muciniphila, the relationship of this bacterium with inflammation development as a predictor of oncogenesis, the influence of other representatives of the intestinal microbiota on its function, and also describe one of the possible mechanisms linking the mucin-degraging ability of this bacterium with the development of oncogenesis.


2020 ◽  
Vol 1 (2) ◽  
pp. 11
Author(s):  
Muhammad Luthfi Adnan

ABSTRAK   Kanker kolorektal merupakan salah satu penyakit keganasan yang paling umum di dunia saat ini. Kanker kolorektal merupakan kasus keganasan paling umum ketiga setelah kanker paru-paru dan payudara serta penyebab kematian paling banyak keempat di dunia. Mwskipun penyebab kanker kolorektal belum diktehaui, namun beberapa faktor risiko seperti faktor familial dapat meningkat risiko terkena kanker kolorektal. Penatalaksaan kanker kolorektal sampai saat ini berupa kemoterapi, radioterapi dan terapi bedah. Pemilihan dari terapi tersebut berdasarkan penilaian kolonoskopi, pemeriksaan radiologi barium dengan teknik kontras ganda, CT colonography dan evaluasi histologi. Studi literatur yang dilakukan menunjukkan adanya ekspresi berlebihan dari reseptor histamin dan adanya defisiensi histamin yang memengaruhi terjadinya kanker kolorektal. Bakteri probiotik Lactobacillus reuteri memiliki efek anti kanker dengan memproduksi histamin yang tidak dapat diproduksi sel kanker, sehingga berperan sebagai antiinflamasi. Buah kurma (Phoenix dactylifera) untuk mendukung aktifitas bakteri probiotik dan memiliki efek anti kanker dalam kandungan buah kurma. Penggunaan bakteri probiotik Lactobacillus reuteri dengan sari buah kurma memiliki efek anti kanker kolorektal sehingga berpotensi sebagai terapi dalam pengobatan kanker kolorektal. Kata Kunci: Buah Kurma, Kanker Kolorektal, Lactobacillus reuteri, Probiotik, Terapi     ABSTRACT   Colorectal cancer is one of the most common malignancies in the world today. Colorectal cancer is the most common malignancy case after lung cancer and cancer as well as the biggest cause of death in the world. Although the cause of colorectal cancer has not been recognized, some risk factors such as family factors can increase the risk of colorectal cancer. Management of colorectal cancer to date is chemotherapy, radiotherapy and surgical therapy. Selection of colonoscopy, barium radiological examination with multiple contrast techniques, CT colonography and histological evaluation. The literature study conducted shows that there are excessive differences in histamine receptors and the presence of histamine deficiencies that affect the increase in colorectal cancer. The probiotic bacteria Lactobacillus reuteri has an anti-cancer effect by producing histamine which cannot be produced by cancer cells, so it works as an anti-inflammatory. Dates (Phoenix dactylifera) to support the activity of probiotic bacteria and have an anti-cancer effect in the content of dates. The use of probiotic bacteria Lactobacillus reuteri with date palm juice has an anti-colorectal cancer effect that is needed as a therapy in the treatment of colorectal cancer. Keywords: Date Fruit, Colorectal Cancer, Lactobacillus reuteri, Probiotics, Therapy


2020 ◽  
Vol 21 (10) ◽  
pp. 3684 ◽  
Author(s):  
Antja-Voy Hartley ◽  
Benlian Wang ◽  
Guanglong Jiang ◽  
Han Wei ◽  
Mengyao Sun ◽  
...  

The overexpression of PRMT5 is highly correlated to poor clinical outcomes for colorectal cancer (CRC) patients. Importantly, our previous work demonstrated that PRMT5 overexpression could substantially augment activation of the nuclear factor kappa B (NF-κB) via methylation of arginine 30 (R30) on its p65 subunit, while knockdown of PRMT5 showed the opposite effect. However, the precise mechanisms governing this PRMT5/NF-κB axis are still largely unknown. Here, we report a novel finding that PRMT5 is phosphorylated on serine 15 (S15) in response to interleukin-1β (IL-1β) stimulation. Interestingly, we identified for the first time that the oncogenic kinase, PKCι could catalyze this phosphorylation event. Overexpression of the serine-to-alanine mutant of PRMT5 (S15A), in either HEK293 cells or CRC cells HT29, DLD1, and HCT116 attenuated NF-κB transactivation compared to WT-PRMT5, confirming that S15 phosphorylation is critical for the activation of NF-κB by PRMT5. Furthermore, the S15A mutant when compared to WT-PRMT5, could downregulate a subset of IL-1β-inducible NF-κB-target genes which correlated with attenuated promoter occupancy of p65 at its target genes. Additionally, the S15A mutant reduced IL-1β-induced methyltransferase activity of PRMT5 and disrupted the interaction of PRMT5 with p65. Furthermore, our data indicate that blockade of PKCι-regulated PRMT5-mediated activation of NF-κB was likely through phosphorylation of PRMT5 at S15. Finally, inhibition of PKCι or overexpression of the S15A mutant attenuated the growth, migratory, and colony-forming abilities of CRC cells compared to the WT-PRMT5. Collectively, we have identified a novel PKCι/PRMT5/NF-κB signaling axis, suggesting that pharmacological disruption of this pivotal axis could serve as the basis for new anti-cancer therapeutics.


2008 ◽  
Vol 36 (06) ◽  
pp. 1019-1028 ◽  
Author(s):  
Chong-Zhi Wang ◽  
Chun-Su Yuan

Colorectal cancer remains one of the most prevalent cancer and a leading cause of cancer related death in the US. Many currently used chemotherapeutic agents are derived from botanicals. Identifying herbal sources, including those from ginseng family, to develop better anti-cancer therapies remains an essential step in advancing the treatment of the cancer. In this article, potential roles of ginseng herbs, especially American ginseng and notoginseng, in colorectal cancer therapeutics are presented. The major pharmacologically active constituents of ginsengs are ginsenosides, which can be mainly classified as protopanaxadiol and protopanaxatriol groups. Structure-activity relationship between their chemical structures and pharmacological activities are discussed. In addition, various steaming temperature and time treatment of the ginseng herbs can change ginsenoside profiles, and enhance their anti-cancer activities. This heat treatment process may increase the role of ginseng in treating colorectal cancer.


2011 ◽  
Vol 49 (05) ◽  
Author(s):  
I Hritz ◽  
Z Varga ◽  
M Juhász ◽  
P Miheller ◽  
Z Tulassay ◽  
...  

2012 ◽  
pp. 84-89
Author(s):  
Quoc Hung Vo ◽  
Nguyen Phuong Nhi Doan ◽  
Dinh Quynh Phu Nguyen ◽  
Thi Dieu Tram Ho ◽  
Thi Hoai Nguyen

Objectives: Nowadays, bioactive substances isolated from marine organisms which are abundant and varied in Vietnamese sea attracted more and more the attention of scientists in the world and Vietnam as well. We have studied on soft coral Sinularia cruciata – Alcyoniidae, which has never been studied in Vietnam before, to find substances which are useful in medical field, especially in anti-cancer therapy. Materials and method: Specimens of soft coral Sinularia cruciata were collected from Con Co, Quang Tri province in May 2011. Pure compounds were isolated by using Thin Layer Chromatography; Column Chromatography normal phase and inverse phase; Shephadex LH 20. Structures of them were determined by spectral data of Nuclear Magnetic Resonance (NMR), Electrospray Ionization Mass Spectrometry (ESI-MS). Results & Conclusion: Structures of 4 compounds were identified: (1) 5.8-epidioxycholest-6-en-3-ol (2) Cholesterol (3) 1-O-hexadecyl-glycerol (Chimyl alcohol) (4) Glycerol 1-O-octadecyl ether (Batyl alcohol). The substance (1) was demonstrated to have strong anti-cancer effects in previous study. Key words Sinularia cruciata, Alcyoniidae, 5,8-epidioxycholest-6-en-3-ol, soft coral, cancer.


Sign in / Sign up

Export Citation Format

Share Document