-
Categories
-
Pharmaceutical Intermediates
-
Active Pharmaceutical Ingredients
-
Food Additives
- Industrial Coatings
- Agrochemicals
- Dyes and Pigments
- Surfactant
- Flavors and Fragrances
- Chemical Reagents
- Catalyst and Auxiliary
- Natural Products
- Inorganic Chemistry
-
Organic Chemistry
-
Biochemical Engineering
- Analytical Chemistry
-
Cosmetic Ingredient
- Water Treatment Chemical
-
Pharmaceutical Intermediates
Promotion
ECHEMI Mall
Wholesale
Weekly Price
Exhibition
News
-
Trade Service
The cancer immune cycle (CIC) includes a series of immune-mediated events required to control tumor growth
.
Blocking one or more steps of CIC can allow tumors to evade immune surveillance
However, attempts to restore anti-tumor immunity by reactivating CIC have so far had limited success
.
In recent years, a large number of studies have shown that the metabolic reprogramming of tumors and immune cells in the tumor microenvironment (TME) is a key factor in immune escape
.
Therefore, the author believes that changes in cell metabolism during tumorigenesis promote the initiation and destruction of CIC
.
The authors also provide a theoretical basis for the metabolic targeting of TME, which may help improve tumor responsiveness to chimeric antigen receptor (CAR) transduced T cells or immune checkpoint block (ICB) therapy
.
Image source: https://pubmed.
ncbi.
The best anti-cancer immune response requires a series of events, collectively referred to as CIC
.
This cycle begins with the release of cancer-associated antigens (CAAs) from dead cancer cells
In mammals, these antigens are captured and processed by dendritic cells (DCs), and then presented to naïve T cells in the tumor-rowing lymph nodes
.
The activated tumor antigen-specific CD8+ T cells mobilize and infiltrate the tumor
They recognize and destroy cancer cells by recognizing homologous peptide antigens (pmc) bound to MHC class I molecules on the surface of cancer cells
.
Subsequently, more CAAs are released to initiate a new round of CIC, and the intensity of the immune response is enhanced in each subsequent round
.
Interference with one or more CIC events can allow tumors to escape immune-mediated destruction, which is a hallmark of cancer
Generally speaking, tumors interfere with CIC by reducing immunogenicity or inhibiting the tumor's osmotic effect on T cells
.
Therapies aimed at reversing these adaptive mechanisms by synthesizing car-transduced T cells or restoring the effector capacity of CD8+ T cells through ICB have led to a landmark clinical trial
However, most patients have a small or short response to immunotherapy, which indicates that the tumor has interrupted CIC at multiple points
.
This article proposes that changes in tumor metabolism are the basic driving factors for tumor immunogenicity and immune evasion, and explores how targeted tumor metabolism can restore functional CIC and promote durable anti-tumor immunity
In order to activate the host immune response, tumors must produce and release abnormal peptides in an environment that activates DC function and appropriate lymph node activation
.
Due to environmental mutations, such as lung cancer or melanoma, or damage to endogenous DNA repair pathways, such as mismatch repair (MMR)-deficient tumors, it has been proposed that sufficient tumor immunogenicity requires a high mutation rate
.
In addition, the authors believe that metabolic reorganization is necessary for tumor growth, which fundamentally leads to tumor immunogenicity
.
Here, the author describes how the metabolic changes that accompany transformation constitute the driving factors of the initial phase of CIC
How does the activation of the oncogenic pathway that drive the growth of cancer cells promote immunogenicity? It has now been determined that growth factor-independent nutrient uptake exceeding the need for ATP production is a sign of tumorigenesis
.
In fact, the high glucose uptake rate is the basis of tumor positron emission tomography (PET) based on 18-fluorodeoxyglucose (FDG) uptake
.
The increase in nutrient intake during tumorigenesis is not limited to glucose, as evidenced by the high intake of glutamine in most tumors
.
Growth factor-independent nutrient uptake is usually driven by the oncogenic activation of phosphoinositide-3 kinase (PI3K) and the MYC pathway, which is one of the most common changes in human cancers
.
How does an increase in nutrient intake lead to mutations? Oncogene-driven nutrient intake leads to the accumulation of mitochondrial reactive oxygen species (ROS), combined with chromatin remodeling, which can lead to an increase in the mutation rate
.
Therefore, the inability to neutralize the oncogene-dependent ROS leads to the activation of cell senescence and the secretion of inflammatory factors, thereby initiating an immune response
.
This has been supported by many studies, including in vitro models.
Normal human fibroblasts overexpress carcinogenic MYC and RAS variants to induce ros-dependent cellular senescence in acute myeloid leukemia, chronic myeloid monocytic leukemia, and juvenile myeloid leukemia.
In monocytic leukemia, ROS induced by kras can increase the secretion of IL-1β in mice and myeloid cells
.
On the contrary, by promoting the inactivation of kelch-like ECH-related protein 1 (KEAP1) or the mutation of nuclear factor erythroid 2 related factor 2 (NRF2) to stabilize the tumor’s inherent antioxidant program, the tumor can be made insensitive to ICB
.
This was confirmed by analyzing the transcriptome data of non-small cell lung cancer (NSCLC) patients treated with the anti-programmed cell death ligand 1 (PD-L1) monoclonal antibody atezolizumab
.
Therefore, the increase in nutrient intake driven by oncogenes can promote mutations, thereby enhancing immunogenicity by increasing the accumulation of ROS in certain cancer cells
.
Tumor immunogenic metabolic regulation model: tumorigenic metabolic reorganization required for tumor cell growth can promote tumor immunogenicity
.
Image source: https://pubmed.
ncbi.
nlm.
nih.
gov/34610889/
The in-depth understanding of how TME enhances immunogenicity and immune evasion through metabolic reprogramming provides some potential new strategies for enhancing anti-tumor immunity
.
This article focuses on how the metabolic reorganization of specific cell types such as tumor cells, DCs, CD8+ T cells, and treg regulate tumor immunogenicity and inhibit anti-tumor immune function
.
In CIC, the author covers various metabolic pathways that play a prominent role in the functions of these cell types, additional metabolic pathways, such as lipid uptake, synthesis and metabolism, and additional stress response pathways, including endoplasmic reticulum stress.
Excited
.
Regarding the stimulation method, whether there is a CIC key modulator is worthy of further exploration
.
In addition, how the metabolic reprogramming of additional non-malignant cells (such as CAFs, endothelial cells, TAMs and MDSCs) in TME regulates CIC is an important research area that may reveal additional therapeutic targets
.
These may include the immunosuppressive metabolic enzymes described herein, which are usually expressed in TME by stromal cells
.
In addition, the impact of cellular metabolic reprogramming on other events within CIC, such as T cell recruitment and tumor infiltration, may provide strategies to reverse the so-called T cell rejection phenotype observed in a variety of immunotherapy refractory tumors
.
Further research on cell type and tumor type specific metabolic reprogramming and how to promote the remodeling of extracellular TME metabolism is a fruitful research field
.
The author believes that a deeper understanding of how tumor cell-specific metabolic reprogramming supports immune evasion will be the key to clinically preventing immune checkpoint inhibitor resistance
.
Finally, extensive analysis of the metabolic network of patients treated with CAR-T cells or ICB can expand the identification of hypothetical targets for combined use and ideally improve the patient's response to such treatments
.
references
Luis F.
Somarribas Patterson et al.
Metabolic regulation of the cancer-immunity cycle Luis F.
Somarribas Patterson.
Trends Immunol 2021 Oct 2;S1471-4906(21)00178-2.
doi: 10.
1016/j.
it.
2021.
09.
002.