Weekly Digests
‹ Back to May

There’s just something about them: predicting response to CAR T cell therapy

May 23, 2018

CD19-directed CAR T cell therapy is a revolutionary tool in the fight against leukemias, however, in patients with chronic lymphocytic leukemia only a small subset benefits. In order to better predict which patients are likely to respond to therapy, and to explore strategies that might expand the portion of responding patients, Fraietta et al. analyzed the T cell intrinsic properties of 41 patients with advanced, heavily pretreated, high-risk CLL who received at least one dose of CD19-directed CAR T cells.

Patients were categorized as complete responders (CR), partial responders who had highly active T cell product and responded to treatment, but relapsed with CLL that had transformed into aggressive B cell lymphoma (PRTD), partial responders (PR), and non-responders (NR). Responding patients (CR/PRTD) exhibited dramatic in vivo expansion of CAR T cells that proved to be essential for antitumor efficacy, and exhibited long-term persistence of functional CAR T cells. To determine whether the distinction between responses might have to do with the intrinsic potency of the CAR T cell product (as opposed to disease type or burden), Fraietta et al. delivered CAR T cells generated from patients to mice engrafted with leukemia cells. CAR T cells generated from responders decreased tumor burden and prolonged survival compared to those generated from partial or non-responders. This served as evidence that there was something about the patient-specific T cell product that could predict antitumor efficacy.

The next step for researchers was determining what those intrinsic factors might be, so they compared the transcriptomes of the CAR T cell infusion products and found that engineered T cells from responders were enriched for expression of genes involved in early memory differentiation. T cells from non-responders, on the other hand, had expression profiles enriched for regulators of late memory, effector T cell differentiation, apoptosis, and aerobic glycolysis that contributes to hypoxia and exhaustion. The researchers demonstrated that inhibition of glycolysis could promote the formation of memory CTL019 lymphocytes with enhanced proliferative capacity after serial restimulation. Upon stimulation, T cells from CR/PRTD patients exhibited a more robust increase in T cell activation genes.

Next, the researchers analyzed cytokines and chemokines produced by CD19-directed CAR T cells and found that those from CR showed higher levels of STAT3 signaling mediators, including serum IL-6, suggesting that STAT3 activation in CAR T cells might be involved in the generation of the potent, less-differentiated T lymphocyte populations. Blockade of the IL-6/STAT3 pathway diminished cells’ proliferative capacity upon serial restimulation, indicating the functional importance of this pathway for expansion and long-term survival after infusion.

Investigating whether certain patient cell populations could predict response to CAR T cell therapy, Fraietta et al. found that the frequency of CD45RO-CD27+CD8+ T cell subset (composed of a unique population of memory cells which are antigen-experienced, persist in a resting state, and possess properties of long-lived memory cells) was significantly higher at the time of leukapheresis in patients who were ultimately responders. This biomarker profile was able to stratify an independent cohort of 8 patients into CR and NR patients with a high degree of specificity. An elevated frequency of cells in this subset was further associated with sustained remission, and accordingly, they were found to divide extensively in culture while maintaining an extended undifferentiated phenotype. When this T cell subset was enriched and used to generate CAR T cells, they potently lysed targets, showed elevated granzyme B production, and expressed lower levels of Ki-67.

Testing whether exhaustion markers might also affect CAR T cell potency, the researchers found that while expanded CAR T cell products from CR patients had lower percentages of PD-1+CD8+ CAR T cells, the presence of exhaustion markers at the time of leukapheresis did not appear to impact future outcomes. Interestingly though, the presence of PD-1- cells within the CD27+CD8+ T cell population within the expanded CAR product strongly segregated responders and nonresponders, and the infusion of a T cell product that contained high doses of PD-1-CD27+CD8+ CAR T cells was highly significantly associated with the likelihood of response to therapy. This subset was further found to mediate tumor control in mice, and was found to have upregulated pSTAT3 in response to IL-6 stimulation. Serum IL-6 may also be a biomarker of potency and persistence of CAR T cell product.

The ability to predict whether a patient will respond to CAR T cell therapy could be used to spare patients from unnecessary costs, side effects, and wasted time. Furthermore, understanding what predicts a successful treatment could be used to develop strategies to select for or enhance certain cellular phenotypes and expand the pool of patients who will benefit from therapy. If the predictive biomarkers identified here hold up in larger cohorts, this tool could become critical in the clinic.

by Lauren Hitchings

References:

Fraietta J.A., Lacey S.F., Orlando E.J., Pruteanu-Malinici I., Gohil M., Lundh S., Boesteanu A.C., Wang Y., O'Connor R.S., Hwang W.T., Pequignot E., Ambrose D.E., Zhang C., Wilcox N., Bedoya F., Dorfmeier C., Chen F., Tian L., Parakandi H., Gupta M., Young R.M., Johnson F.B., Kulikovskaya I., Liu L., Xu J., Kassim S.H., Davis M.M., Levine B.L., Frey N.V., Siegel D.L., Huang A.C., Wherry E.J., Bitter H., Brogdon J.L., Porter D.L., June C.H., Melenhorst J.J. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nat Med. 2018 May.

In the Spotlight...

A CD19/CD3 bispecific antibody for effective immunotherapy of chronic lymphocytic leukemia in the ibrutinib era

Robinson et al. developed a CD19/CD3 bispecific antibody in the single-chain Fv-Fc format with a half-life of approximately 5 days. In human ex vivo and murine xenograft in vivo models of chronic lymphocytic leukemia (CLL), the antibody induced autologous T cell expansion, activation, and granzyme B expression, and led to robust CLL cell killing in blood and spleen. The antibody was effective with PBMCs from treatment-naive and ibrutinib-treated patients, and induced a faster response in the latter case, suggesting synergy with ibrutinib. Response was also observed against CLL cells from patients who were ibrutinib-resistant.

CSF1R+ Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype

After showing that CSF1R expression in human pancreatic ductal adenocarcinoma (PDAC) is associated with poor prognosis, Candido et al. inhibited CSF1R phosphorylation in mice with pancreatic cancer using the small molecule AZD7507. AZD7507 monotherapy led to the depletion of CSF1R+PD-L1+ tumor-associated macrophages, looser stroma, reduction in pro-tumor cytokines and chemokines, increased infiltration of CD8+ and CD4+ T cells (but not Tregs), reduced tumor mass, and prolonged survival. AZD7507 altered the PDAC gene expression away from the squamous subtype, which is associated with poor outcome.

Revealing the specificity of regulatory T cells in murine autoimmune diabetes

Using a GFP reporter construct, Spence et al. demonstrate that Tregs within the inflamed islets of Langerhans in nonobese diabetic (NOD) mice are locally stimulated and activated by islet antigens via the TCR. Deep TCR sequencing showed clonal expansion and accumulation within the islets. Single cell paired αβTCR sequencing reveals that a significant portion of islet Tregs is insulin-specific, and an adoptive transfer of islet Tregs into NOD.C28-/- mice efficiently prevents diabetes development, pointing to the use of insulin-specific TCRs for engineered Treg therapy.

Cross-talk between T cells and hematopoietic stem cells during adoptive cellular therapy for malignant glioma

Wildes et al. found that concurrent adoptive transfer of tumor-reactive T cells and bone marrow-derived hematopoietic stem and progenitor cells (HSPCs) into irradiated mice with glioblastoma, brain stem glioma, or medulloblastoma led to the differentiation of intracranial HSPCs (recruited via the CXCL12-CXCR4 axis) into dendritic cells (DCs), reduction in myeloid-derived suppressor cells, prolonged survival, and tumor rejection. In a feedback loop, IFNγ released by activated antigen-specific T cells led to HSPC differentiation into DCs, which further promoted T cell activation.

Novel effector phenotype of Tim-3+ regulatory T cells leads to enhanced suppressive function in head and neck cancer patients

Liu et al. showed that Tim-3+ Tregs in TILs from head and neck squamous cell carcinomas were more suppressive in vitro than their Tim-3- counterparts, and they aimed to understand the mechanism. Tim-3+ Tregs expressed higher levels of co-inhibitory molecules (PD-1 and CTLA-4), granzyme B (but not higher IL-10 or LAP), and IFN-γ receptor. In vitro, IFN-γ or anti-PD-1 treatment reduced Tim-3+ Treg suppressive activity. After five treatments in an in vivo murine model, anti-PD-1 treatment decreased Tim-3 expression, suggesting a possible contribution of Treg suppression to anti-PD-1 therapy.

Immune Profiling of Premalignant Lesions in Patients With Lynch Syndrome

To characterize the immune profile of premalignant lesions, or polyps, in patients with the mismatch repair (MMR)-associated disease, Lynch Syndrome (LS), Chang et al. performed RNA sequencing on polyps from patients with LS and the non-MMR disease, familial adenomatous polyposis. Their analysis revealed an immune activated profile (CD4+ T cell infiltration, proinflammatory molecules, and checkpoint molecules) in LS premalignant polyps that was independent of mutational rates, neoantigen formation, or mismatch repair status, challenging the current paradigm.

M7824, a novel bifunctional anti-PD-L1/TGFbeta Trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine

Knudson et al. tested M7824, a fusion protein that combines TGFβRII with anti-PD-L1, in murine primary and metastatic models of breast and colon carcinoma. M7824 targeted to the tumor via PD-L1 binding, sequestered TGFβ, and activated NK and CD8+ T cells, leading to reduced tumor growth, increased median overall survival (OS), and memory response in mice with complete tumor rejection. The antitumor efficacy was dependent on NK and CD8+ T cells. M7824 combined with a tumor-associated antigen TWIST1 vaccine further increased OS; unexpectedly, combining vaccine with anti-PD-L1 abrogated the antitumor effect of anti-PD-L1.

Oncolytic influenza virus infection restores immunocompetence of lung tumor-associated alveolar macrophages

Studying infection with oncolytic influenza A virus (IAV) in an immunocompetent Raf-BxB spontaneous NSCLC murine model, Masemann et al. observed that infection with IAV not only directly decreased the number and size of lung tumors via viral lytic growth, but also restored the pro-inflammatory and anti-tumoral effects of previously tumor-suppressed alveolar macrophages in the lungs in vivo. Infection caused a shift in the tissue resident M2-like macrophage to an iNOS+, MHCII+ anti-tumor phenotype.

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.