1. Resources
  2. Articles
  3. Targeting the Tumor microenvironment 丨How to turn "Cold" Tumors become "hot" ?

Targeting the Tumor microenvironment 丨How to turn "Cold" Tumors become "hot" ?
Tumor microenvironment (TME) is the internal environment for tumor cell growth and progression. In contrast to traditional thinking, TME is considered a complex integrated system, composed of cellular components such as tumor cells and inflammatory/immune cells, as well as non-cellular components such as extracellular matrix (ECM) and cytokines . During cancer development,"TME is not just a silent bystander, but rather an active promoter of cancer progression".
"Hot" and "cold" tumors

Based on the growth characteristics, tumors can be divided into "malignant" and "benign". The difference between "hot" and "cold" tumors will be explained in the following paragraphs.

According to the spatial distribution of immune cells in TME, tumors can be classified into three basic immunophenotypes: immune-inflamed, immune-excluded, and immune-desert phenotypes.

"Hot tumors", also known as Immune-inflamed tumors, are characterized by high T-cell infiltration, increased interferon-γ (IFN-γ) signaling, expression of PD-L1, and high tumor mutational burden (TMB) . (In general, tumors with a higher TMB carry a higher neoantigen load that can be recognized by T cells, making them more likely to prime the immune system). CD8+ T lymphocytes play roles in the prolonged survival of cancer patients and increased efficacy of immunotherapy. Infiltration of CD8+ T lymphocytes is critical for cancer immunotherapies. Immune-excluded and immune-desert tumors can be described as "cold tumors". In immune-excluded tumors, CD8+ T lymphocytes localize only at invasion margins and do not efficiently infiltrate the tumor. In immune-desert tumors, CD8+ T lymphocytes are absent from the tumor and its periphery.

Figure 1. Differentiation of 'hot and cold' tumors based on the distribution of immune cells
Figure 1. Differentiation of "hot and cold" tumors based on the distribution of immune cells[1].
Based on the spatial distribution of CD8+ T lymphocytes in the TME, a gradient of three immunophenotypes is observed: the immune-desert, immune-excluded and immune-inflamed phenotypes.

Immunosuppressive mechanisms of the TME in cold tumors: Besides poor T cell infiltration, the T cell priming may be inhibited in cold tumor TME. Furthermore, the deposition of extracellular matrix (ECM) and stiff stroma-induced hypoxia in cold tumor lesions build a physical and chemical barrier to obstruct the T cell infiltration. More notably, numerous immunosuppressive cells are widely settled in the TME of cold tumors and suppress the function of CD8+ T cells via T cell exhaustion[2](Figure 2). In summary, TME in cold tumors lacks innate immunity, or "cold tumors" possess innate anti-tumor immunity characteristics.

Figure2. Immunosuppressive mechanisms of the tumor immune microenvironment in cold tumors
Figure2. Immunosuppressive mechanisms of the tumor immune microenvironment in cold tumors
When hot and cold tumors hit immune checkpoints

Immune checkpoints are the inhibitory pathways on the surface of T cells that inhibit immune activation. Immune checkpoint Inhibitors (ICIs) have revolutionized cancer treatment by activating T-cell-based antitumor immunity. The TME in hot tumors is infiltrated by numerous T cells, and the immune cells in "hot" TME tumors are more active than in cold tumors. When ICIs remove the inhibitory effect of the immune checkpoint, immune response of T cells can be activated again, then the effector T cells kill the cancer cell. "Hot tumors" with inflammatory phenotypes tend to be more sensitive to ICIs therapy; however, ICIs are not effective against cold tumors due to their "stiff" and immunosuppressive microenvironment.

Therefore, conversion of "cold tumors" to "hot tumors" has become an attractive strategy for cancer treatment, which could increase the sensitivity ofthese tumors to ICIs and improveclinical use ofimmunotherapy

How do you turn a cold tumor into a hot tumor

Driven T cells into the cold tumor TME improve the efficacy of ICI

STING agonists, oncolytic viruses, chemotherapy, radiotherapy, etc.can induce immunogenic cell death (ICD) to promote T-cell priming and activation[1][3].

For example, activation of the STING signaling pathway mediates the expression of proinflammatory cytokines (e.g., type I IFNs) and activates the innate immune responses mediated by T cells.The activated immune system changes the immunophenotype of the tumor.It switches the "cold tumor" with low T cell reactivity to the "hot tumor" with high T cell reactivity.

As illustrated in figure 3, In the article entitled Development of Potent Immune Modulators Targeting Stimulator of Interferon Genes Receptor, STING agonist 4c significantly decreased tumor volume in a CT26 murine colorectal carcinoma model. Additionally, In a 4C-treated mouse model, immunological memory-derived cancer inhibition was observed. The results confirm the therapeutic potential of compound 4c for cancer immunotherapy via STING-mediated immune activation[3]. In line with the above observatione, an article published in Science also reported that MSA-2 (a STING agonist) stimulates the secretion of IFN-β and induces tumor regression. In addition, MSA-2 increases CD8+ T lymphocyte infiltration in low PD-1 blocking response mouse tumor models.

Figure 3. Sting-mediated immune activation achieves cancer immunotherapy
Figure 3. Sting-mediated immune activation achieves cancer immunotherapy

Increase the abundance of immune checkpoints and promote effective T cells infiltration,"assisting" cold tumors turn to hot

In March 2022, Dr. Jinfang Zhang's team at Wuhan University published their study entitled "USP8 inhibition reshapes an inflamed tumor microenvironment that potentiates the latest achievement in the field of tumor immunotherapy". The study reveals that inhibition of the deubiquitination enzyme USP8 can reshape the tumor immune microenvironment (TME), and turn "cold" tumors to "hot", thereby improving an inflamed TME where immunotherapy can be more effective.

The article shows that in vitro inhibition of USP8 can enhance the ubiquitination modification of PD-L1 K63 junction and up-regulate PD-L1 protein level. Additionally, a combination of USP8 inhibitor with anti-PD-L1 antibody significantly suppressed tumor growth and improved the overall survival rates of MC38 tumor-bearing C57BL/6 mice(Fig 4a). A combination of DUB-IN-2 and anti-PD-L1 antibodies could significantly increase the percentage of CD8+ T cells (Fig. 4b). Meanwhile, USP8 inhibitor combined with anti-PD-L1 treatment also significantly elevated the expression of the T cell activation marker GzmB and reduced the expression of the exhausted T cell marker TIM3 on infiltrated CD8+ T cells (Fig.4c-d). This study provides evidence to support the enhanced anti-tumor efficacy of the combined therapies of a USP8 inhibitor and PD-1/PDL-1 blockade.

Figure 4. USP8 reshapes the Tumor Immune Microenvironment (TME)
Figure 4. USP8 reshapes the Tumor Immune Microenvironment (TME)[4].
a-b Tumor growth or KaplanMeier survival curves for C57BL/6 bearing MC38 tumors. c-d Quantification of T cells in CT26 tumors. e-f. Quantification of GzmB or TIM3 on TILs in CT26 tumors

The nanoparticle-based treatment for turning "cold" tumors into "hot"

Nanotechnologies are rapidly increasing their role in immunotherapy in line with the need for novel therapeutic strategies. Nanoparticle (NP)-based drugs for turning “cold tumors” into “hot tumors”.Four potential pathways in Fig 5 show the mechanisms of NP-based drugs turning cold tumors hot[5]:

Figure 5: Nanoparticle-based treatment for turning cold tumors
Figure 5: Nanoparticle-based treatment for turning cold tumors[5].
(1) NP-Mediated shaping of physical barriers and target of stromal cells, using nanocarriers to improve tumor targeting and protect the payload from degradation within the TME, promoting better penetration of immune cells and therapies; (2) Using NPS-coated siRNA to reduce the expression of crucial proteins and inhibit cell invasion and migration; (3) Targeting cytokine signals and elevating immune response represents a valuable strategy to turn a cold tumor hot; (4) NPs induce cell stress and immunogenic cell death. Figure 5: Nanoparticle-based treatment to warm cold tumors[5]
Summary

1.The difference between "hot" and "cold" tumors is mainly due to the spatial distribution of immune cells, and "cold" tumors are often "insensitive" to immunotherapy.

2.Promoting T cell initiation could produce by increasing tumor antigen expression and restoring antigen processing and presentation mechanisms (e.g., STING agonist[3]). Furthermore, reprogramming the tumor microenvironment, promoting T-cell transportation and allowing T-cells to infiltrate effectively[4]. These methods potentially could turn "cold" tumors to "hot" and therefore improve immunotherapy efficiency.

Related Products

DUB-IN-2

A potent deubiquitinase inhibitor with an IC50 of 0.28 μM for USP8.

ML364

A selective USP2 inhibitor with anti-proliferative activity, which direct binds to USP2 (Kd=5.2 μM).

P 22077

A cell-permeable ubiquitin-specific protease 7 (USP7) inhibitor with an EC50 of 8.01 μM.

PR-619

A broad-range and reversible DUB inhibitor . PR-619 induces ER Stress and ER-Stress related apoptosis.

MSA-2

A potent and orally available non-nucleotide STING agonist, is bound to STING as a noncovalent dimer with nanomolar affinity.

ADU-S100 disodium salt

An activator of stimulator of interferon genes (STING).

Ubiquitination Compound Library

A unique collection of 200+ small molecule modulators with biological activity used for ubiquitination research. Compounds in this library target the key enzymes in ubiquitin pathway.

Immunology/Inflammation Compound Library

A unique collection of 3893 compounds that are useful tool for Immunology/Inflammation research or autoimmune inflammatory diseases drug discovery.