Weekly Digests
‹ Back to June

CD4+ CAR T cells kill tumor cells from a distance with IFNγ

June 14, 2023

CD8+ T cells often take center stage when it comes to immunotherapy, but in some cases, CD4+ T cells contribute strongly, and may even exert more potent effects than CD8+ T cells. Investigating the variable contributions of CD4+ T cells to antitumor efficacy in CAR T cell therapy, Boulch et al. explored the various functionalities of CAR CD4+ T cells (CAR4 T cells) and found that while they do kill some tumor cells directly through perforin/granzyme B, they primarily induce indirect tumor cell killing through IFNγ. The results were recently published in Nature Cancer.

The researchers began by identifying mouse models that were sensitive (pro-B-cell model) and insensitive (Eμ-myc model) to CAR4 T cell-mediated tumor control. To track tumor cell death, they performed intravital imaging on the bone marrow of mice with tumors expressing a caspase 3 reporter, and as expected, caspase 3 activity/cell death was much higher in the CAR4 T cell-treated pro-B cell model compared to the Eu-myc model.

To parse out how CAR4 T cells induce tumor cell death, mice bearing pro-B-cell tumors were treated with either wild-type (WT) or IFNγ-/- CAR4 T cells. While WT CAR4 T cells mediated tumor control and a survival advantage, these effects were greatly diminished, and the survival advantage was lost when IFNγ production was knocked out. Intravital imaging confirmed that IFNγ-/- CAR4 T induced lower levels of caspase 3 activity in tumor cells.

Next, the researchers used intravital imaging again to classify tumor cell killing events as direct or indirect, depending on whether they occurred with or without detectable CAR T cell contact. Interestingly, less than a third of CAR4 T cell-induced cell deaths were associated with direct contact, while the majority of deaths were indirect. The majority of indirect cell killing (and some direct cell killing) was lost with the use of IFNγ−/− CAR4 T cells, while most direct cell killing was lost with the use of Prf1−/− CAR4 T cells, suggesting that IFNγ mediates killing of tumor cells from a distance, while perforin/granzyme B mediates most killing at close range. Importantly, while loss of perforin/granzyme and direct cell killing had little impact on the efficacy of CAR4 T cells, loss of IFNγ was accompanied by a loss of therapeutic benefit, suggesting that this mechanism of tumor cell killing plays a dominant role in mediating tumor control.

Next, the researchers found that CAR4 T cells, but not IFNγ−/− CAR4 T cells, substantially raised serum IFNγ in WT hosts, and controlled pro-B-cell tumors in IFNγ−/− hosts, suggesting that host-derived IFNγ does not play a major role in the therapeutic activity of CAR4 T cells. Further, while CAR8 T cells could independently control pro-B-cell tumors (mainly via the perforin pathway), they did little to increase IFNγ.

To assess the dynamics of IFNγ diffusion in tumors, the researchers utilized a fluorescent reporter for STAT1 activity, and tracked it using intravital imaging to reveal that both direct injection of recombinant IFNγ and transfer of CAR4 (but not CTRL4) T cells resulted in STAT1 translocation throughout tumors, suggesting efficient cytokine diffusion. At tumor sites, CAR4 T cell therapy led to upregulation of MHC-I, ICAM-1, and PD-L1 (all classically upregulated by IFNs) in both tumor and immune cells at the tumor site.

Investigating exactly how CAR4 T cells and IFNγ mediate long-distance killing, the researchers found that it was not due to indirect effects on other effector cells, nor due to an impact on other host cells, as results were similar across WT, Prf1-/-, and IFNγR1-/- mice. Instead, the effects were found to be due to direct IFNγ-mediated killing, as pro-B-cell tumors underwent apoptosis upon exposure to IFNγ in vitro, and were controlled by direct administration of IFNγ in vivo in both WT and IFNγR1−/− mice. Meanwhile, CAR4 T cells were ineffective against IFNγR1−/− tumors, highlighting the importance of IFNγ sensing by tumor cells for antitumor efficacy. The capacity for long-distance killing was further tested and validated using Transwell assays with both mouse and human cells.

Given the capacity of CAR4 T cells to kill at a distance via IFNγ, Boulch et al. evaluated whether CAR4 T cells could effectively target antigen escape variants. In mixed samples of CD19+ and CD19- pro-B cells, the researchers observed bystander killing of CD19- targets. Similar results were observed with human cells and in a mixed CD19+/CD19- tumor model in vivo.

Different tumors are known to exhibit different sensitivities to IFNγ-induced cell death, and Boulch et al. demonstrated this using several cell lines, including solid tumor lines. Among various pro-B-cell lines (newly generated by viral Abl overexpression), all responded to IFNγ exposure by upregulating MHC-I and PD-L1, but only some showed substantial cell death. When mice were administered tumors containing a mix of fluorescently labeled cells from 3 IFNγ-sensitive and 1 IFNγ-insensitive pro-B-cell line, only the sensitive cell lines were efficiently controlled by CAR4 T cells (but not IFNγ-/- CAR4 T cells), suggesting that their efficacy was highly dependent on tumor sensitivity to IFNγ-mediated killing. Differential tumor sensitivity did not appear to be linked to IFNγR expression, suggesting that other tumor-intrinsic factors likely play a role in determining sensitivity.

Finally, in data from a cohort of 63 patients with diffuse large B-cell lymphoma who were treated with CAR T cells, the researchers monitored CAR4:CAR8 T cell ratios in the blood and IFNγ concentrations in serum. At 1 week post-transfer, around the peak of CAR T cell expansion, CD4:CD8 T cell ratios varied extensively between patients, and patients with high (above median) CAR4:CAR8 T cell ratios exhibited stronger induction of IFNγ. Among patients with high (but not low) CAR4:CAR8 T cell ratios, high IFNγ induction (above the median) was associated with significantly improved progression-free and overall survival.

Overall, Boulch et al. showed that CAR4 T cells primarily exert antitumor effects through production of IFNγ, which can diffuse throughout tumors to induce apoptosis in distant tumor cells that are sensitive to IFNγ-mediated killing. Moving forward, understanding the contributions of these mechanisms and the sensitivity of different tumors to IFNγ could help to predict and/or improve patient responses to CAR4 T cells.

Write-up and image by Lauren Hitchings.

Meet the Researcher

This week, first author Morgane Boulch answered our questions.

First author Morgane Boulch

What was the most surprising finding of this study for you?
We observed that CD4+ CAR T cells kill tumor cells in an unexpected manner, at distance from tumors, without physically contacting them, but by secreting IFNγ. In the lab, we rely on intravital imaging, a non-biased approach that allows us to explore the mode of action of different immunotherapies in real time. In the past, we have observed that CD8+ CAR T cells, thought to be the main effector cells, engaged into direct cellular contacts with tumors to induce cell death. It turns out to be quite different for CD4+ T cells, and I still remember the day when I first saw tumor apoptotic events popping up at distance from CD4+ CAR T cells.

What is the outlook?
We showed that in lymphoma patients with a high CD4:CD8 CAR T cell ratio, high IFNγ serum level was associated with a better survival. This observation, with the demonstration that, in preclinical models, IFNγ can limit the emergence of antigen escape variants, suggest that it might be helpful to design CAR-based strategies that maximize IFNγ production. In the future, it will also be important to define the optimal CAR T cell product composition (especially the CD4:CD8 ratio) for each individual tumor, in particular, based on its sensitivity to cytokine-induced cell death.

What was the coolest thing you’ve learned (about) recently outside of work?
Two weeks ago, while I was visiting a vineyard and testing wine in Burgundy (France), I found out that French vineyards almost entirely disappeared in the 19th century in France because of a tiny insect called Phylloxera vastatrix. That was until the discovery in the late 1870s that grafting the European fruit-bearing vine variety Vitis vinifera on the root of an American vine, naturally adapted and immune to Phylloxera, protects the sensitive European vine from the Phylloxera attack. Today, almost all European vines are grafted onto American rootstocks. To me, this story is a perfect illustration of innovation, adaptation and collaboration – values shared within the scientific community.

References:

Boulch M, Cazaux M, Cuffel A, Guerin MV, Garcia Z, Alonso R, Lemaître F, Beer A, Corre B, Menger L, Grandjean CL, Morin F, Thieblemont C, Caillat-Zucman S, Bousso P. Tumor-intrinsic sensitivity to the pro-apoptotic effects of IFN-γ is a major determinant of CD4+ CAR T-cell antitumor activity. Nat Cancer. 2023 May 29.

In the Spotlight...

Leukemia relapse via genetic immune escape after allogeneic hematopoietic cell transplantation

Among 494 patients with myeloid neoplasms treated with HSCT, patients with high class-II HLA evolutionary divergence (HED, an indirect measure of epitope diversity) had a lower rate of relapse and better overall survival than those with low class-II HED. High class-I and -II HED was also associated with an increased number, but low clonal expansion of cancer-related clonotypes. In post-transplant relapses (but not relapse directly following chemotherapy), HLA-I and -II genes were preferentially targeted by somatic mutations, or expression was downregulated, suggesting HLA mutation and dysregulation as mechanisms of immune escape.

Contributed by Lauren Hitchings

Gene-edited and -engineered stem cell platform drives immunotherapy for brain metastatic melanomas

Kanaya et al. showed that PTEN loss was associated with melanoma brain metastasis (MBM) and poor prognosis through immunosuppressive mechanisms, and established and characterized immunosuppressive PTEN-deficient MBM mouse models. Delivery of oncolytic herpes simplex virus (oHSV) via infected stem cells (SCs) showed immunogenic cell death of tumor cells and therapeutic effects in vitro and in vivo. Locoregional delivery of oHSV-resistant nectin-1 receptor knockout SCs engineered to deliver GM-CSF and an anti-PD-1 scFv boosted SC-oHSV efficacy and systemic antitumor T cell immunity in both syngeneic and patient-derived PTEN-deficient MBM mice.

Contributed by Shishir Pant

Cell surface marker-based capture of neoantigen-reactive CD8+ T-cell receptors from metastatic tumor digests

By using flow cytometry to sort T cells co-expressing PD-1, CD39, and TIGIT (TILTP) from tumor digests of metastatic epithelial cancers, Chatani et al. were able to detect and enrich neoantigen-reactive CD8+ T cells. Their results were comparable to those achieved with a single-cell transcriptomic approach, but used more readily available and affordable technology. While the sorted TILTP cells were highly enriched for tumor reactivity, they lacked proliferative capacity, and were unable to maintain reactivity through in vitro expansion processes, making them unsuitable as products for ACT. Instead, rapid TCR identification could be utilized in a TCR engineering approach.

Contributed by Lauren Hitchings

Preclinical Evidence of an Allogeneic Dual CD20xCD22 CAR to Target a Broad Spectrum of Patients with B-cell Malignancies

With the goal of improving outcomes and treatment options for patients with relapsed or refractory B-cell lymphoma, Aranda-Orgilles et al. generated an allogeneic TRAC-/CD52- dual CD20x22 CAR from healthy T cell donors. Preclinically, allogeneic CD20x22 CAR T cells exhibited strong, sustained, and dose-dependent cytolytic activity against tumor cells expressing one or both antigens, and efficiently killed primary human B-NHL samples with variable CD20/CD22 levels. In a patient-derived B-NHL xenograft model, CD20x22 CAR T cells increased overall survival, providing a proof of concept for overcoming resistance mechanisms and offering an option to CD19 targeting.

Contributed by Katherine Turner

Targeted TLR9 Agonist Elicits Effective Antitumor Immunity against Spontaneously Arising Breast Tumors

In the physiologically relevant MMTV-PyMT GEMM model, Miller and Sagiv-Barfi et al. demonstrated that systemic delivery of a tumor integrin-targeted TLR9 agonist (PIP-CpG) led to sustained elimination of tumors existing at the time of treatment (“initial” tumors) and delayed growth, but eventual progression with subsequent newly induced tumors. Effective PIP-CpG therapy depended on CD4+/CD8+ T cells and resulted in profound TME remodeling, affecting cytokine/chemokine production, innate and adaptive immune cell infiltration, and immune cell phenotypic changes, on balance resulting in an antitumor immune landscape.

Contributed by Ed Fritsch

Chronic antigen in solid tumors drives a distinct program of T cell residence

Gavil et al. used parabiosis to characterize the migrational properties of CD8+ T cells in normal murine mammary fat pads (MFP) and orthotopic breast tumors. Conventional TRM markers derived from virus-specific memory CD8+ T cells, such as CD69 and CD103, identified MFP TRM, but failed to correlate with tumor residence. In the tumor context, CD62L, a marker typically associated with circulating non-resident T cells, was also expressed in TRM. Chronic antigen stimulation and the tumor environment drove distinct programs of residence in tumors, whereby tumor-specific stem progenitor CD8+ T cells migrate to tumors and acquire markers of exhaustion while becoming resident.

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.