Weekly Digests
‹ Back to August

Inducing CAR-macrophages in the brain for local treatment

August 17, 2022

Despite aggressive treatment involving debulking surgery and chemoradiotherapy, glioblastoma multiforme (GBM) remains incurable. A major culprit for GBM recurrence and therapy resistance are self-renewing glioma stem cells (GSCs) that are left behind after surgery. Additionally, immunosuppressive macrophages are recruited to the tumor cavity in response to surgery-induced inflammation. Chen and Jing et al. investigated whether these intracavitary macrophages could be redirected to induce phagocytic activity against GSCs by engineering them to express GSC-specific chimeric antigen receptors (CARs). Their data describing the efficacy of this approach in animal models were recently published in Science Translational Medicine.

Using bioinformatics, the researchers found that CD133 expression was high in glioma tissues, and variable expression levels were detected on patient-derived glioblastoma cells. High CD133 expression was correlated with tumor recurrence and poor survival. Therefore, the researchers constructed macrophage-specific promoter-driven anti-CD133 CAR plasmids (pCAR) encoding the CD3ζ intracellular costimulatory domain. Macrophage-targeted pCAR-laden nanoporters (NP-pCAR) were synthesized with cationic peptides and a brain extracellular matrix (ECM)-mimetic hydrogel was created to serve as the transporter for NP intracavitary immobilization. The NP-pCAR released from the hydrogel was designed to target the macrophages in the cavity and introduce the CD133-specific CAR into the macrophages' nuclei.

The researchers designed a positively amphipathic chimera for nuclear-targeted gene delivery using a nuclear localization sequence peptide as the hydrophilic moiety and palmitic acid as the hydrophobic part. The amphipathic PA-peptide self-assembled into a nanomicelle, into which the negatively charged pCAR was loaded through electrostatic interactions. The pCAR-laden nanomicelle was then further coated with citraconic anhydride-modified dextran to produce a CD206 receptor-targeted pCAR-laden nanomicelle (NP).

To test the efficiency of the NP-mediated gene delivery, the researchers made use of THP-1 cells, bone marrow-derived macrophages (BMDM), and RAW 264.7 cells. In these macrophages, the cellular integration of genes was enhanced when they were encapsulated in dextran-coated NP in vitro. The NP efficiently delivered the genes into macrophages with the dextran-targeting motif; the NP escaped the lysosome and entered the cytosol. It was also shown that the macrophages induced functional CAR expression and the macrophages had increased phagocytic activity against CD133+ cells.

Proteomics analysis of the CAR-macrophages showed that M1-associated pathways were induced. Several M1 macrophage-associated biomarkers were upregulated, while M2 proteomic markers were inhibited. There was an increase in the expression of CD80, iNOS, TNFα, and IL-1β, while ARG-1 and IL-10 expression were reduced. These data suggest that the NP-pCAR could introduce the anti-CD133 CAR into macrophages and shift their phenotype to the classically proinflammatory M1 subtype.

The effects of this strategy on antitumor responses was tested in the GL261 syngeneic orthotopic mouse glioma model. After tumors were established, these were injected with gene-laden NP (pCAR dose 2 mg/kg). On day 6 after treatment, the CAR-macrophage frequency among all F4/80+ cells was approximately 5%, which increased to about 12% on day 12. In vivo phagocytic cell killing of CD133-expressing tumor cells by NP-pCAR-treated macrophages was observed. Treatment with NP-pCAR or anti-CD47 antibody alone reduced tumor burden, while the combination treatment induced the most tumor regression. The combination treatment group had the highest ratio of healthy brain tissue versus tumor areas. This resulted in an 83% survival rate at day 120, while this combination did not induce severe organ damage or body weight loss.

Long-term survivors in the combination treatment group were rechallenged with glioma cells in the opposite hemisphere to assess whether the treatment had induced immunological memory. The mice remained tumor-free and survived long-term. In these mice, there was an increase in the percentage of effector memory and central memory T cells in the blood and spleen. The mice also had higher levels of serum cytokines, including IFNγ, IL-12, IL-15, and TNFα. These data suggest that the combination treatment induced adaptive immune responses.

To assess the treatment in a situation that would occur in patients after surgery, the researchers made use of a brain tumor resection mouse model, in which 12 days after tumor inoculation with GL261 cells, the visible tumor was surgically removed under a stereoscope. In the tissue surrounding the resection cavity, remaining tumor cells and macrophages were observed. Then, to test the treatment, the cavity was injected with hydrogel formulations containing NP-CARs and/or anti-CD47 antibody. In mice treated with the NP-pCAR gel, there was a decrease in tumor cell bioluminescence signals on day 33 after inoculation. The efficacy was even higher when this treatment was combined with an anti-CD47 antibody, and mice survived until at least day 120. No toxicity was detected in macrophages, neurons, or astrocytes, and there was no sign of gliosis. In the combination treatment group, the tissue surrounding the cavity on day 19 after inoculation contained higher levels of CD8+ T cells and M1 macrophages and lower levels of M2 macrophages than control groups. Furthermore, secretion of IFNγ, IL-12, and TNFα was elevated in the combination group.

To assess the CAR gene delivery efficiency in the human setting, human THP-1 cells and patient-derived glioma (PDG) xenograft models were used. The monocytic THP-1 cells were induced with an M2-like phenotype and treated with the NP-pCAR. More than 31% of cells had detectable EGFP (a fluorescent indicator for CAR transduction). These cells had enhanced phagocytotic activity against tumor cells and upregulated secretion of TNFα and IL-1β, and there was an increase in the frequency of CD80+ M1 macrophages. Finally, an orthotopic GBM model was established, in which PDG cells were intracranially injected into human hematopoietic stem cells NOD.Cg-Prkdcscid Il2rgtm1SugTg(SV40/HTLV-IL3,CSF2)10-7Jic/Jic (HuHSC-NOG-EXL) mice. After the surgical debulking of established tumors, the hydrogel-supported combination treatment was injected into the cranial cavity. This induced tumor growth inhibition, and 83% of mice survived more than 90 days, suggesting this method is also effective in humanized models.

The data presented here reveal a new approach to treating GBM to reduce tumor recurrence after debulking surgery. If these data can be repeated in patients, the retargeting of macrophages and the local induction of adaptive immune responses might improve patient outcomes.

Write up by Maartje Wouters, image by Lauren Hitchings.

References:

Chen C, Jing W, Chen Y, Wang G, Abdalla M, Gao L, Han M, Shi C, Li A, Sun P, Jiang X, Yang Z, Zhang S, Zhang J, Tang C, Liu Y, Zhang R, Xu F, Dong B, Li X, Liu M, Qiang B, Sun Y, Wei X, Li J, Hu Q, Jiang X. Intracavity generation of glioma stem cell-specific CAR macrophages primes locoregional immunity for postoperative glioblastoma therapy. Sci Transl Med. 2022 Aug 3.

In the Spotlight...

An anti-CTLA-4 heavy chain-only antibody with enhanced T(reg) depletion shows excellent preclinical efficacy and safety profile

Gan et al. created a human CTLA-4-specific fully human heavy chain-only antibody (4003-1), which bound with high affinity to a CTLA-4 epitope overlapping with that bound by ipilimumab, and which similarly inhibited CTLA-4 binding to B7-1 and B7-2. In vitro, an IgG1Fc-mutated version (4003-2) more potently induced T cell activity, partly due to enhanced Treg depletion. In human CTLA-4 knock-in mouse tumor models, 4003-2 efficiently penetrated tumors and depleted Tregs, and had potent antitumor efficacy and a shorter systemic half life. In a 3-month, multidose, cynomolgus monkey study, 4003-2 had low systemic drug exposure, anticipated pharmacodynamics, and good tolerability.

Contributed by Paula Hochman

Cell-penetrating peptides enhance peptide vaccine accumulation and persistence in lymph nodes to drive immunogenicity

Backlund and Holden et al. demonstrated that conjugates of antigen and cell-penetrating peptides (CPP) promoted dendritic cell-mediated antigen uptake and enhanced peptide vaccine potency in vivo. Antigen–CPP conjugates primed a polyfunctional T cell population in a cross-presenting DC-dependent manner, but not via cytosolic delivery of peptide to the MHC class I antigen processing pathway. Linkage to CPPs significantly enhanced antigen accumulation in draining lymph nodes (dLNs), increased uptake by APCs in dLNs, protected linked antigens from degradation in serum, and prolonged the duration of antigen presentation in dLNs.

Contributed by Shishir Pant

Cis interactions between CD2 and its ligands on T cells are required for T cell activation

Li et al. generated CD2 or CD48 (murine CD2 ligand) KO mice and tagged-CD2 knockin mice. CD2 KO or Ab blockade, or CD48 KO blunted in vitro TCR-stimulated T cell responses in the presence or absence of CD48+ APCs, while CD2-dependent T cell-mediated cytolysis in vitro partially required CD48+ APCs. GVHD was reduced by the absence of CD48, but more so CD2, on grafted T cells. FRET imaging and cell mixing showed CD2–CD48 cis interactions on individual T cells. T cell activation mediated via CD2 depended on CD2 cytoplasmic domain proline-rich motifs, which bound lck in vitro. Human CD2–CD58 (LFA-3, human CD2 ligand) cis interactions also promoted human TCR signaling.

Contributed by Paula Hochman

Dual-antigen targeted off-the-shelf NK cells show durable response and prevent antigen escape in lymphoma and leukemia

Cichocki et al. generated human iPSC-derived NK cells expressing a CD19-CAR containing the NKG2D, 2B4, and CD3ζ signaling domains, a high-affinity, non-cleavable CD16 shown to enhance therapeutic Ab-directed ADCC, and a membrane-bound IL-15/IL-15R fusion protein shown to boost general and CAR-directed NK cytolysis and in vivo persistence. These CAR NK cells had a propensity to kill malignant over healthy CD19+ cells and, unlike CD19-CAR T cells, in settings mimicking antigen escape, killed CD19+ and CD19- human B cell tumors. Their activity was enhanced by rituximab. In xenogeneic adoptive transfer models, these CAR NK cells eliminated CD19+ and CD19- lymphoma cells.

Contributed by Paula Hochman

Size-dependent activation of CAR-T cells

Xiao et al. tested how CAR T cell activation is affected by the relative lengths of the CAR construct, the antigen target, and the membrane phosphatase CD45. Reducing the length of the CAR extracellular domain (by removing Ig domains) enhanced exclusion of CD45 from the CAR synapse, increased CAR T cell activation (intracellular phosphorylation, cytotoxicity, and cytokine production), and better restrained tumor growth in vivo. Decreasing antigen length or increasing the size of CD45 (mAb binding or a longer isoform) had similar effects, indicating that CAR T cell activation and antitumor efficacy are enhanced by exclusion of CD45 from the CAR synapse, and can be physically tuned.

Contributed by Alex Najibi

Transcriptomic diversity in human medullary thymic epithelial cells

Carter et al. combined high-throughput 5’Cap sequencing with standard RNA sequencing to investigate gene expression and transcript diversity in immature (MHC-IIlo) and mature (MHC-IIhi) human mTECs, and demonstrated increased rates of global transcript mis-initiation among the mature mTEC population. In the mature mTEC population, AIRE-dependent, but not FEZF2-dependent, genes showed increased rates for transcript mis-initiation. Differential expression of peripheral splicing factors drove alternative splicing in both mTECs, whereas, during mTEC maturation, specific EREs enriched in long terminal repeat retrotransposons were upregulated.

Contributed by Shishir Pant

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.